CN117321046A - Indazole-based compounds and related methods of use - Google Patents

Indazole-based compounds and related methods of use Download PDF

Info

Publication number
CN117321046A
CN117321046A CN202280035511.XA CN202280035511A CN117321046A CN 117321046 A CN117321046 A CN 117321046A CN 202280035511 A CN202280035511 A CN 202280035511A CN 117321046 A CN117321046 A CN 117321046A
Authority
CN
China
Prior art keywords
alkyl
group
optionally substituted
ptm
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280035511.XA
Other languages
Chinese (zh)
Inventor
埃里卡·阿劳乔
迈克尔·柏林
史蒂文·M·斯帕克斯
王静
张伟
董汉清
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Avenas Enterprises
Original Assignee
Avenas Enterprises
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Avenas Enterprises filed Critical Avenas Enterprises
Priority claimed from PCT/US2022/021049 external-priority patent/WO2022198112A1/en
Publication of CN117321046A publication Critical patent/CN117321046A/en
Pending legal-status Critical Current

Links

Abstract

A bifunctional compound of formula PTM-L-CLM, wherein CLM has the formula al-a4, PTM has the formula PTM-IA and L is a ligand. These compounds are useful in treating cancer by agonism of LRRK2 and cereblon E3 ubiquitin ligaseDry disease plays a role.

Description

Indazole-based compounds and related methods of use
Cross Reference to Related Applications
The present disclosure claims priority and equity from U.S. provisional application No. 63/163,328, filed on 3 months, 19, 2021, U.S. provisional application No. 63/228,731, filed on 8 months, 3, 2021, and U.S. provisional application No. 63/243,014, filed on 9, 10, 2021, and U.S. provisional application No. 63/245,411, filed on 9, 17, 2021, each entitled "INDAZOLE BASED COMPOUNDS AND ASSOCIATED METHODS OF USE" and incorporated herein by reference in its entirety.
Incorporated by reference
All cited references are hereby incorporated by reference in their entirety, including U.S. patent application Ser. No. 17/207,325, published under the heading "INDAZOLE BASED COMPOUNDS AND ASSOCIATED METHODS OF USE" filed on day 19, 3, 2021, U.S. patent application publication No. 2021/0315896, and U.S. patent application Ser. No. 14/686,640, published on day 14, 2015, 4; and U.S. patent application Ser. No. 14/792,414, published as U.S. patent application publication 2016/0058872, filed on 7/6 of 2015; and U.S. patent application Ser. No. 15/953,108, published as U.S. patent application publication No. 2018/0228907, filed on day 13 of 4 in 2018; and U.S. patent application publication 2016/0009689A1 filed on 9/2/2015; and U.S. patent application publication 2016/0200722A1 filed 18 at month 2 of 2016.
Technical Field
The present invention provides heterobifunctional compounds comprising a target protein binding moiety and an E3 ubiquitin ligase binding moiety and related methods of use. The bifunctional compounds are useful as modulators of targeted ubiquitination of leucine-rich repeat kinase 2 (LRRK 2), which leucine-rich repeat kinase 2 (LRRK 2) is then degraded and/or inhibited.
Background
Most small molecule drugs bind enzymes or receptors in tight and well-defined pockets. On the other hand, it is well known that protein-protein interactions are difficult to target with small molecules due to their large contact surface, shallow grooves involved, or flat interfaces. E3 ubiquitin ligases, hundreds of which are known in humans, confer ubiquitination specificity to substrates and, therefore, due to their specificity for certain protein substrates, they are more attractive therapeutic targets than general proteasome inhibitors. The development of E3 ligase ligands has proven challenging, in part because they must disrupt protein-protein interactions. However, recent developments provide specific ligands that bind to these ligases. For example, since the discovery of the first small molecule E3 ligase inhibitor nutlin, additional compounds targeting E3 ligase have been reported.
Cereblon is a protein encoded by the CRBN gene in humans. CRBN orthologs are highly conserved from plant to human, emphasizing their physiological importance. Cereblon forms an E3 ubiquitin ligase complex with modulators of damaged DNA binding protein 1 (DDB 1), cullin-4A (CUL 4A) and Cullin 1 (ROC 1). This complex ubiquitinates many other proteins. By a mechanism that has not been fully elucidated, cereblon ubiquitination of target proteins results in elevated levels of fibroblast growth factor 8 (FGF 8) and fibroblast growth factor 10 (FGF 10). FGF8 in turn regulates many developmental processes such as limb and auditory vesicle formation. The end result is that this ubiquitin ligase complex is important for limb growth within the embryo. In the absence of cereblon, DDB1 forms a complex with DDB2, which acts as a DNA-damage binding protein.
Bifunctional compounds, such as those described in U.S. patent application publications 2015/0291562 and 2014/0356322 (incorporated herein by reference), function to recruit endogenous proteins to the E3 ubiquitin ligase for ubiquitination and subsequent degradation in the proteasome degradation pathway. In particular, the publications cited above describe bifunctional or proteolytically targeted chimerism Protein degrading agent compounds which can be used as modulators of targeted ubiquitination of a variety of polypeptides and proteins which are then degraded and/or inhibited by the bifunctional compounds.
Leucine rich repeat kinase 2 (LRRK 2) is a member of the leucine rich repeat kinase family and is a large multi-domain protein with an N-terminal armadillo domain, an ankyrin repeat region, a Leucine Rich Repeat (LRR) domain, a tandem Roco gtpase domain, a kinase domain containing a DFG-like motif and a C-terminal WD40 domain. The LRRK2 protein has 2527 amino acids and a molecular weight of 280kDa. The catalytic activity of LRRK2 is related to the kinase and GTPase domains, and the active form of LRRK2 is a heterodimer (Greggio E et al: the Parkinson disease-associated leucine-rich repeat kinase 2 (LRRK 2) is a dimer that undergoes intramolecular autophosphorylation. Jbiol Chem 2008,283: 16906-16914). GTP binding is essential for kinase activity, and mutations that prevent GTP binding have been shown to eliminate LRRK2 kinase activity (Ito G et al: GTP binding is essential to the protein kinase activity ofLRRK2, acausative gene product for familial Parkinson's disease. Biochemistry2007, 46:1380-1388). The only validated physiological substrates (excluding LRRK2 itself) are a subset of low molecular weight G proteins, including Rab8a and Rab10, which are involved in regulating vesicle transport and endosomal function and transport on the cytoskeletal network (Steger M et al Phosphoproteomics reveals that Parkinson's disease kinase LRRK2regulates a subset of Rab GTPases. Elife 2016,5. E12813). LRRK2 is expressed at the highest level in immune cells (neutrophils, monocytes and B cells), lung and kidney, and at lower levels in the brain (which is expressed in dopaminergic neurons of the substantia nigra) (West AB et al Differential LRRK2 expression in the cortex, stratum, and substantia nigra in transgenic and nontransgenic logs j Comp neurol2014, 522:2465-2480).
LRRK2 has several major functionally acquired pathogenic and characteristic mutations located in the Roco domain (N1437H, R1441G/C/H, Y1699C) to effect GTP hydrolysis or in the kinase domain (G2019S and I2020T). G2019S is the most common LRRK2 mutation associated with parkinson' S disease (PD), a progressive neurodegenerative disorder characterized by resting tremor, rigidity, hypokinesia (bradykinesia) and postural instability. The histological features of PD include neurodegeneration of dopaminergic neurons in the substantia nigra pars compacta, intracellular inclusion bodies called lewy bodies, and neurites consisting of aggregated forms of α -synuclein. G2019S is associated with 1% -2% of all PD patients and results in a 2-fold increase in kinase activity in vitro (West AB et al: parkinson' S diseaseassociated mutations in leucine-rich repeat kinase 2augment kinase activity.Proc Natl Acad Sci U S A2005,102:16842-16847) and a 4-fold increase in autophosphorylation at Ser1292 (Sheng Z et al: ser1292 autophosphorylation is an indicator of LRRK2 kinase activity and contributes to the cellular effects of PD mutations. Sci Transl Med 2012,4: 164ra161). The G2019S and I2020T mutations are located within the DFG motif (DYGI in the case of LRRK 2) and are common to all kinases, which control catalytic activity. These mutations are believed to disrupt the inactive conformation and thereby increase catalytic activity (Schmidt SH et al: the dynamic switch mechanism that leads to activation of LRRK2 is embedded in the DFGpsi motif in the kinase domain. Proc Natl Acad Sci USA 2019,116: 14979-14988). Several of the above Parkinson's disease-associated mutations (R1441C/G, Y1699C and I2020T) inhibit the phosphorylation of LRRK2 at Ser910 and Ser935, which in turn reduces the association of LRRK2 with 14-3-3 protein, which is believed to represent an inactive form of LRRK2 (Nichols J et al: 14-3-3binding to LRRK2 is disrupted by multiple Parkinson's disease associated mutations and regulates cytoplasmic localisation.Biochem J2010,430:393-404).
In addition, LRRK2 is associated with autosomal dominant inherited PD by a mutation within the region of chromosome 12 designated PARK8, which is associated with the LRRK2 gene (Funayama M et al: A new locus for Parkinson's disease (PARK 8) maps to chromosome p11.2-q13.1.Ann Neurol 2002,51:296-301; zimphch A et al: mutations in LRRK2 house auto-dominant parkinsonism with pleomorphic strategy. Neuron 2004,44:601-607; paisan-Ruiz C et al: cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease. Neuron 2004,44: 595-600). LRRK2 was first described in 1978 as being associated with autosomal dominant inherited parkinson's disease, where it dates back to a family of japan (Nukada H et al: [ A big family of paralysis agitans (author' stransl) ]. Rinsho Shinkeigaku 1978, 18:627-634). The most common pathogenic LRRK2 mutations (G2019S) occur in 4% -8% of familial PD cases and 1% -3% of sporadic PD cases. Furthermore, the G2019S mutation is common in PD patients of selected lineages, with 30% -40% of north africa Bai Baier patients and 14% of jerusalem artichoke patients harboring the mutation.
LRRK2 kinase inhibitors have been proposed to have potential to treat mutation-driven PD (where LRRK2 activity is increased), such as G2019S, and idiopathic PD (where LRRK2 activity is increased) (Chen J et al: leucine-rich repeat kinase 2in Parkinson's disease:updated from pathogenesis to potential therapeutic target.Eur Neurol2018,79:256-265; alessi DR et al: LRRK2 kinase in Parkinson ' S disease.science 2018,360:36-37; di Maio R et al: LRRK2 activation in idiopathic Parkinson ' S disease.Sci Transl Med 2018,10). Several therapeutic agents are entering the clinic, including LRRK2 kinase inhibitors that directly affect downstream target phosphorylation, and oligonucleotides (ASOs) that are infused directly into the CNS to block translation of LRRK2 protein and thereby reduce LRRK2 protein levels.
The lewy body is the main histological marker of PD. The Lewis body consists mainly of aggregates of Alpha-synuclein, and increasing mutations in such aggregated Alpha-synuclein also increases the risk of developing PD (Meade RM et al: alpha-synuclein structure and Parkinson' sdisease lessons and emerging principles. Mol neurogenin 2019,14.29-29). Depletion of LRRK2 with ASO (Zhao HT et al: LRRK2 antisense oligonucleotides ameliorate a-synuclein inclusion formation in aParkinson's disease mouse model. Molecular therapy. Nucleic acids 2017, 8:508-519) and loss of LRRK2 at the genomic level have been shown to reduce alpha-synuclein-mediated lesions in PD mouse models (LinX et al: leucine-rich repeat kinase 2regulates the progression of neuropathology induced by Parkinson's-disease-related mutant alpha-synuclein. Neuron 2009, 64:807-827). Mutations that increase LRRK2 activity, such as G2019S, increase aggregation of α -synuclein in neuronal and PD mouse models. This increase was reversed by LRRK2 kinase inhibitor (Volpicelli-Daley LA et al G2019S-LRRK2 Expression Augments a-Synuclein Sequestration into Inclusions in neurons. JNEurosci.2016, 7 months 13; 36 (28): 7415-27.Doi: 10.1523/JNEROSCI.3642-15.2016). There is some evidence that The G2019S mutant form of LRRK2 is resistant to inhibition by kinase inhibitors in The CNS, potentially reducing its disease-modifying effects (Kelly K et al, the G2019S mutation in LRRK2 imparts resiliency to kinase inhibitor. Exp neuron. 2018nov; 309:1-13). Although most PD cases also had a lewy body at necropsy, no lewy body was present in a large number of PD cases associated with LRRK 2G 2019S mutations (Kalia LV et al: clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease JAMA neuron 2015, 72:100-105). In addition to the fact that lewy bodies are a common feature of PD, tau lesions are also a major feature of LRRK2 mutant carriers at necropsy (Henderson MX et al: alzheimer 'sdisease Tau is a prominent pathology in LRRK2 Parkinson's disease. Acta Neuropathol Commun 2019,7.183-183). In one Study, tau lesions were observed in 100% of LRRK2 mutant carriers, understating that LRRK2 is an important target for linking PD to Tau lesions in the case of PD, although genetic causal relationships were not as strong as between LRRK2 and primary tauopathies such as supranuclear palsy (PSP) or corticobasal degeneration (CBD) (Ross OA et al (2006) LRRK 2R 1441 substitution and progressive supranuclear palsy. Neuro Appl neuro 32 (1): 23-25; sanchez-control M et al (2017) Study of LRRK2variation in tauopathy: progressive supranuclear palsy and corticobasal degeneration. Mov dis 32 (1): 115-123). A common variation at the LRRK2 locus was recently reported as a genetic determinant of PSP survival (Jabbari E et al Common variation at the LRRK2 locus is associated with survival in the primary tauopathy progressive supranuclear palsy. BioRxiv 2020.02.04.932335; doi: https:// doi.org/10.1101/2020.02.04.932335). Increased expression of LRRK2 in PSP obtained by analysis of the expression quantitative trait locus (eQTL) was reported to potentially lead to reactive microglial-induced pro-inflammatory states that drive sustained accumulation of misfolded Tau protein and clinical disease progression. Functional variants of LRRK2 are also associated with Crohn's disease and leprosy type 1 inflammatory responses (Hui KY et al Functional variants in the LRRK2 gene confer shared effects on risk for Crohn's disease and Parkinson's disease. Sci Transl Med.2018, 10; 10 (423): sai 7795.Doi:10.1126/scitranslmed. Aai7795; fava et al Pleiotropic effects for Parkin and LRRK in leprosy type-1reactions and Parkinson'sdisease.Proc Natl Acad Sci U S A.2019, 7, 30; 116 (31): 15616-15624.Doi: 10.1073/pnas.1905116. Epub 2019,7, 15).
LRRK2 is highly expressed in the immune system in neutrophils, monocytes and macrophages, and brain microglia, and is a regulator of the intrinsic regulation of microglial activation and lysosomal degradation processes (Ma et al Genetic comorbidities in Parkinson's disease.hum Mol genet.2014, month 2, 1; 23 (3): 831-41.doi:10.1093/hmg/ddt465.epub 2013, month 9, 20, reviewed in Schapansky et al The complex relationships between microglia, alpha-synuclein, and LRRK2 in parkin's disease.neuroscience.2015, month 27; 302:74-88.doi:10.1016/j.neuroscience.2014.09.049, 2014, month 10, month 2). Prolonged activation of these immune cells by PD disease processes or mutations in LRRK2 may increase neuroinflammation and lead to a greater risk of developing PD and/or Tau lesions. Treatment with an Anti-TNF agent reduced the risk of developing PD in patients with inflammatory bowel disease by 78% (Peter I et al: anti-tumor necrosis factor therapy and incidence of Parkinson disease among patients with inflammatory bowel disease.jama neuron 2018), demonstrating a close link between inflammation and PD. In addition to PD, LRRK2 is associated with other diseases such as cancer, leprosy and Crohn's disease (Lewis PA, manzoni C.LRRK2and human disease: a complicated question or a question of complexes (2012). Sci Signal.5 (207), pe 2).
There is a continuing need in the art for effective treatments for LRRK 2-associated diseases and disorders such as idiopathic PD, LRRK2 mutation-associated PD (e.g., PD associated with one or more LRRK2 activating mutations), primary tauopathies (e.g., supranuclear palsy (PSP) or corticobasal degeneration (CBD)), lewy body dementia, crohn's disease, leprosy (e.g., leprosy with type 1 inflammatory response), and/or neuroinflammation.
Disclosure of Invention
The present disclosure describes heterobifunctional compounds for recruiting leucine-rich repeat kinase 2 (LRRK 2) to E3 ubiquitin ligase for targeted ubiquitination and subsequent proteasome degradation, and methods of making and using the heterobifunctional compounds. Furthermore, the present disclosure provides methods of using an effective amount of a compound of the present disclosure to treat or ameliorate a disease condition, such as an LRRK 2-related disease or disorder, e.g., accumulation or overactivity of LRRK2 protein or mutated LRRK2 protein or misfolded LRRK2 protein, or α -synuclein accumulation or accumulation, or Tau accumulation or idiopathic PD, or LRRK2 mutation-related PD (e.g., PD associated with one or more LRRK2 activating mutations), or primary tauopathies (e.g., supranuclear palsy (PSP) or corticobasal degeneration (CBD)), or lewy body dementia, or crohn's disease, or leprosy (e.g., leprosy with type 1 inflammatory response), or neuroinflammation.
Thus, in one aspect, the present disclosure provides heterobifunctional compounds comprising an E3 ubiquitin ligase binding moiety (i.e., a ligand of an E3 ubiquitin ligase ("ULM" group)) and a moiety that binds LRRK2 or a mutant form thereof (i.e., a protein targeting moiety or "PTM" group, i.e., a ligand or "LTM" group that targets LRRK 2) such that LRRK2 protein is thereby placed in proximity to ubiquitin ligase to effect ubiquitination and subsequent degradation (and/or inhibition) of LRRK2 protein. In a preferred embodiment, the ULM (ubiquitinated ligase binding moiety) is a cereblon E3 ubiquitin ligase binding moiety (CLM). For example, the structure of a bifunctional compound can be depicted as:
the respective positions of the PTM and ULM portions (e.g., CLM), and the amounts thereof as exemplified herein, are provided by way of example only and are not intended to limit the compounds in any way. As the skilled artisan will appreciate, bifunctional compounds as described herein may be synthesized such that the number and location of the individual functional moieties may be varied as desired.
In certain embodiments, the bifunctional compound further comprises a chemical linker ("L"). In this example, the structure of the bifunctional compound can be depicted as:
Where PTM is an LRRK2 targeting moiety (LTM), L is a linker, e.g., a bond or chemical linking group coupling PTM to ULM, and ULM is a cereblon E3 ubiquitin ligase binding moiety (CLM).
For example, the structure of a bifunctional compound can be depicted as:
wherein: PTM is LRRK2 targeting moiety (LTM); "L" is a linker (e.g., a bond or a chemical linking group) coupling PTM to the CLM; and CLM is a cereblon E3 ubiquitin ligase binding moiety that binds to cereblon.
In any aspect or embodiment described herein, PTM is a small molecule that binds LRRK2 or a mutant thereof. In any aspect or embodiment described herein, PTM is a small molecule that binds LRRK2. In any aspect or embodiment described herein, PTM is a small molecule that binds to LRRK2 wild-type protein and LRRK2 mutant (such as LRRK2 mutant comprising one or more mutations selected from G2019S, I2020T, N1437H, R1441G/C/H and Y1699C). In any aspect or embodiment described herein, PTM is a small molecule that binds to LRRK2 wild-type protein and LRRK2 mutant (such as but not limited to G2019S, I2020T, N1437H, R1441G/C/H, Y1699C or a combination thereof). In any aspect or embodiment described herein, the small molecule binds LRRK2 as described herein.
In one embodiment, the CLM comprises chemical groups derived from imides, thioimides, amides, or thioamides. In particular embodiments, the chemical group is a phthalimido group, or an analog or derivative thereof. In a certain embodiment, the CLM is selected from thalidomide (thalidomide), lenalidomide (lenalidomide), pomalidomide (pomalidomide), their analogs, their isosteres, and their derivatives. Other contemplated CLMs are described in U.S. patent application publication No. 2015/0291562, which is incorporated by reference herein in its entirety.
In certain embodiments, "L" is a bond. In further embodiments, the linker "L" is a linker having a linear number of non-hydrogen atoms ranging from 1 to 20. The linker "L" may include, but is not limited to, one or more functional groups such as ethers, amides, alkanes, alkenes, alkynes, ketones, hydroxy, formic acid, sulfides, sulfoxides, and sulfones. The linker may comprise an aromatic, heteroaromatic, cyclic, bicyclic or tricyclic moiety. Halogen (such as Cl, F, br, and I) substitutions may be included in the linker. In the case of fluorine substitution, a single or multiple fluorine groups may be included.
In certain embodiments, CLM is a derivative of piperidine-2, 6-dione, where the piperidine-2, 6-dione may be substituted at the 3-position, and the 3-substitution may be a bicyclic heteroarene, whose linkage is a C-N bond or a C-C bond. Examples of CLMs may be, but are not limited to pomalidomide, lenalidomide, and thalidomide, and their analogs.
In another aspect, the present description provides therapeutic compositions comprising an effective amount of a compound as described herein, or a salt form thereof, and a pharmaceutically acceptable carrier. The therapeutic compositions may be used to trigger targeted degradation of LRRK2 or a mutant form thereof and/or inhibition of LRRK2 or a mutant form thereof in a patient or subject (e.g., an animal such as a human), and may be used to treat or ameliorate one or more disease states, conditions, or symptoms of the patient or subject that are causally related to LRRK2 or a mutant form thereof by degrading or inhibiting LRRK2 protein or a mutant form thereof, or controlling or reducing the protein level of LRRK2 protein or a mutant form thereof. In certain embodiments, therapeutic compositions as described herein may be used to effect degradation of LRRK2 or mutant forms thereof to treat or ameliorate diseases such as, for example, LRRK2 accumulation or hyper-reactivity, α -synuclein accumulation or accumulation, tau accumulation or accumulation, idiopathic PD, LRRK2 mutation-related PD (e.g., PD associated with one or more LRRK2 activating mutations), primary tauopathies (e.g., supranuclear palsy (PSP) or corticobasal gangrene (CBD)), lewy body dementia, crohn's disease, leprosy (e.g., leprosy with type 1 inflammatory response), and/or neuroinflammation.
In yet another aspect, the present disclosure provides a method of ubiquitinating LRRK2 or a mutant form thereof in a cell. In certain embodiments, the methods comprise administering a heterobifunctional compound as described herein comprising a PTM that binds LRRK2 or a mutant form thereof, and CLM, preferably linked by a chemical linker moiety as described or exemplified herein, to effect degradation of LRRK2 protein or a mutant form thereof. While not wanting to be limited by theory, the inventors believe that, in accordance with the present invention, when LRRK2 wild-type or mutant proteins are placed in proximity to E3 ubiquitin ligase by using heterobifunctional compounds, polyubiquitination of LRRK2 wild-type or mutant proteins will occur, triggering subsequent degradation of LRRK2 or mutant proteins via the proteasome pathway and controlling or reducing LRRK2 protein levels in cells (such as cells of a subject in need of such treatment). The control or reduction of the level of LRRK2 protein or mutant forms thereof provided by the present disclosure provides for the treatment of a disease state, disorder or symptom associated with LRRK2 cause and effect, such as by reducing the amount of LRRK2 protein or mutant forms thereof in cells of a subject.
In yet another aspect, the present specification provides a method for treating or ameliorating a disease, disorder, or symptom thereof that is causally related to LRRK2 or a mutant form thereof in a subject or patient (e.g., an animal such as a human), the method comprising administering to a subject in need thereof a composition comprising an effective amount (e.g., a therapeutically effective amount) of a heterobifunctional compound as described herein, or a salt form thereof, and a pharmaceutically acceptable carrier, wherein the composition is effective to treat or ameliorate the disease or disorder, or symptom thereof in the subject.
In another aspect, the present description provides methods for identifying the effect of degrading LRRK2 protein in a biological system using a compound according to the present disclosure.
In another aspect, the present specification provides methods and intermediates for preparing heterobifunctional compounds of the present disclosure that are capable of targeting ubiquitination and degradation of LRRK2 proteins in cells (e.g., in vivo or in vitro).
Drawings
The accompanying drawings, which are incorporated in and form a part of this specification, illustrate several embodiments of the present disclosure and, together with the description, serve to explain the principles of the disclosure. The drawings are only for purposes of illustrating embodiments of the disclosure and are not to be construed as limiting the disclosure. Further objects, features and advantages of the present disclosure will become apparent from the following detailed description taken in conjunction with the accompanying drawings that illustrate exemplary embodiments of the present disclosure.
FIGS. 1A and 1B illustrate the general principle of heterobifunctional protein degrading compounds. FIG. 1A exemplary heterobiofunctional protein degrading compound comprises a protein targeting moiety (PTM; dark shaded rectangle), ubiquitin ligase bindingMoieties (ULM; light shaded triangles) and optionally linker moieties (L; black lines) coupling PTM to ULM. FIG. 1B illustrates a heterobifunctional protein degrading compound as described herein (commercially termed Protein degrading compounds). Briefly, ULM (triangle) recognizes and binds to a specific E3 ubiquitin ligase, while PTM (large rectangle) binds and recruits a target protein so that it is in close proximity to the E3 ubiquitin ligase. Typically, the E3 ubiquitin ligase is complexed with the E2 ubiquitin conjugated protein (E2) and catalyzes the attachment of multiple ubiquitin molecules (black circles) via isopeptide bonds to lysines on the target protein either alone or through the E2 protein. The polyubiquitin protein (rightmost) is thus targeted for degradation by the proteasome mechanism of the cell.
Detailed Description
Presently described are compounds, compositions, and methods relating to the surprising discovery that an E3 ubiquitin ligase (e.g., cereblon E3 ubiquitin ligase) ubiquitinates an LRRK2 protein or mutant form thereof once the E3 ubiquitin ligase and LRRK2 protein are placed in proximity by a bifunctional compound that binds the E3 ubiquitin ligase and the LRRK2 protein. Accordingly, the present disclosure provides compounds and compositions comprising an E3 ubiquitin ligase binding moiety ("ULM") coupled to a protein targeting moiety ("PTM") targeting LRRK2 protein via a bond or chemical linking group (L), which results in ubiquitination of LRRK2 protein and degradation of LRRK2 protein by proteasome (see fig. 1).
In one aspect, the present description provides compounds wherein PTM binds to LRRK2 protein and/or mutant forms thereof. The disclosure also provides libraries of compositions and their use in the targeted degradation of LRRK2 proteins in cells.
In certain aspects, the disclosure provides heterobifunctional compounds comprising a ligand, such as a small molecule ligand (i.e., having a molecular weight of less than 2,000, 1,000, 500, or 200 daltons), that is capable of binding to an E3 ubiquitin ligase, such as cereblon. The compounds also comprise a small molecule moiety capable of binding to LRRK2 protein or a mutant form thereof in such a way that the LRRK2 protein or a mutant form thereof is placed in proximity to ubiquitin ligase to effect ubiquitination and degradation (and/or inhibition) of the LRRK2 protein or a mutant form thereof. By "small molecule" is meant that, in addition to the above, the molecule is non-peptidyl, i.e., it is not considered a peptide, e.g., comprising less than 4, 3, or 2 amino acid residues. According to the present description, each of the PTM, ULM and heterobifunctional molecules is a small molecule.
The term "LRRK2" as used throughout the specification is intended to include both wild-type LRRK2 and mutant forms thereof, such as LRRK2 muteins comprising one or more mutations selected from G2019S, I2020T, N1437H, R1441G/C/H and Y1699C, unless specifically indicated to the contrary.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The terminology used in the description is for the purpose of describing particular embodiments only and is not intended to be limiting of the disclosure.
Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise (such as where a group of multiple carbon atoms is provided for each carbon atom falling within the range), and any other stated or intervening value in that stated range is encompassed within the disclosure. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where a stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the disclosure.
The following terms are used to describe the present disclosure. Where a term is not specifically defined herein, the term is given its art-recognized meaning by the ordinarily skilled artisan, who employ the term in describing the present disclosure in the context of its use.
The articles "a" and "an" as used herein and in the appended claims are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article unless the context clearly dictates otherwise. For example, unless otherwise indicated, "an element" refers to one element or more than one element.
In the claims and the above patent specification, all transitional phrases such as "comprising", "including", "carrying", "having", "containing", "involving", "holding", "consisting of … …", etc. are to be understood as being open, i.e. meant to include but not limited to. As described in section 2111.03 of the U.S. patent office patent review program manual, only the transitional phrases "consisting of … … (confusing of)" and "consisting essentially of … … (consisting essentially of)" should be closed or semi-closed transitional phrases, respectively.
It should also be understood that, in some methods or processes described herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited, unless the context indicates otherwise.
The terms "co-administration" and "co-administration" or "combination therapy" refer to concurrent administration (simultaneous administration of two or more therapeutic agents) and time-varying administration (administration of one or more therapeutic agents at a different time than administration of another therapeutic agent) so long as the two or more therapeutic agents are present in the patient to some extent, preferably in an effective amount at the same time. In certain preferred aspects, one or more heterobifunctional compounds described herein are co-administered with at least one additional bioactive agent (e.g., an anticancer agent). In particularly preferred aspects, co-administration of such compounds results in synergistic activity and/or therapy, such as, for example, anticancer activity.
The term "compound" as used herein refers to any particular heterobifunctional compound disclosed herein, pharmaceutically acceptable salts and solvates thereof, as well as deuterated forms of any of the foregoing molecules, if applicable, unless otherwise indicated. Deuterated compounds are contemplated which have one or more hydrogen atoms contained in the drug molecule replaced with deuterium. Such deuterated compounds preferably have one or more improved pharmacokinetic or pharmacodynamic properties (e.g., longer half-life) as compared to an equivalent "non-deuterated" compound.
The term "ubiquitin ligase" refers to a family of proteins that promote the transfer of one or more ubiquitin to a specific substrate protein. The goal of adding chains of several ubiquitin (polyubiquitination) is to degrade the substrate protein. For example, cereblon is an E3 ubiquitin ligase, alone or in combination with E2 ubiquitin conjugating enzymes, which ultimately can result in the linkage of a chain of four ubiquitin to lysine residues on a target protein, thereby targeting the protein for degradation by the proteasome. Ubiquitin ligases are involved in polyubiquitination such that the first ubiquitin is linked to a lysine on the target protein; the second ubiquitin is linked to the first ubiquitin; the third ubiquitin is linked to the second ubiquitin and the fourth ubiquitin is linked to the third ubiquitin. Such polyubiquitinated tag proteins are degraded by the proteasome.
The term "patient" or "subject" is used throughout the specification to describe an animal, preferably a human or a domesticated animal, to whom treatment (including prophylactic treatment) with the compositions of the present disclosure is provided. For the treatment of a disease, disorder, or symptom that is characteristic of a particular animal (such as a human patient), the term "patient" refers to the particular animal, including domestic animals (such as dogs or cats) or farm animals (such as horses, cattle, sheep, etc.). Generally, in this disclosure, the terms "patient" and "subject" refer to a human patient unless otherwise indicated or implied in the context of use of such terms.
The terms "effective" and "therapeutically effective" are used to describe the amount of a compound or composition that achieves a desired result, such as an improvement in a disease or disorder, or an improvement or reduction in one or more symptoms associated with a disease or disorder, when used in the context of its intended use, as well as after a single dose or more preferably multiple doses in the context of a therapeutic regimen. The terms "effective" and "therapeutically effective" include all other "effective amount" or "effective concentration" terms that are otherwise described or used in this application.
Compounds and compositions
In one aspect, the present description provides heterobifunctional compounds comprising an E3 ubiquitin ligase binding moiety ("ULM"), namely a cereblon E3 ubiquitin ligase binding moiety ("CLM"). CLM is covalently coupled to a Protein Targeting Moiety (PTM) that binds to a protein, said coupling being performed directly by a bond or by a chemical linking group (L) according to the following structure:
(A)PTM-L-CLM
wherein L is a bond or a chemical linking group, and PTM is a protein targeting moiety that binds to protein LRRK2 or a mutant form thereof (e.g., G2019S), wherein PTM is an LRRK2 targeting moiety (LTM). The term CLM includes all cereblon binding moieties.
In any aspect or embodiment, CLM exhibits a half maximal Inhibitory Concentration (IC) of less than about 200 μm for an E3 ubiquitin ligase (e.g., cereblon E3 ubiquitin ligase) 50 )。IC 50 May be determined according to any suitable method known in the art (e.g., fluorescence polarization measurement).
In certain embodiments, the heterobifunctional compounds described herein exhibit an IC of less than about 100, 50, 10, 1, 0.5, 0.1, 0.05, 0.01, 0.005, 0.001mM, or less than about 100, 50, 10, 1, 0.5, 0.1, 0.05, 0.01, 0.005, 0.001 μm, or less than about 100, 50, 10, 1, 0.5, 0.1, 0.05, 0.01, 0.005, 0.001nM, or less than about 100, 50, 10, 1, 0.5, 0.1, 0.05, 0.01, 0.005, 0.001pM 50 Or half maximum Degradation Concentration (DC) 50 )。
In any aspect or embodiment described herein, the heterobifunctional compound is represented by the following chemical structure:
/>
wherein:
Z 1 an R group of a CLM as described in any aspect or embodiment herein modified to be covalently linked to L, such group selected from a bond, -C (=o) -, -CONR '-, -O-, -NR' -, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
R is selected from bond, H, O, OH, N, NH, NH 2 -Cl, -F, -Br, -I, methyl, optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl), optionally substituted linear or branched alkoxy (e.g., optionally substituted linear or branched C1-C6 alkoxy), -alkyl-aryl (e.g., an-alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxyl; and is also provided with
Q 1 、Q 2 、Q 3 、Q 4 、Q 5 、R 4 、L、R 1 、R 2 、M、X 1 、X 2 、X 3 、X 4 、X 5 、X 6 、X 7 、X 8 Andas defined in any aspect or embodiment described herein.
In any aspect or embodiment described herein, the heterobifunctional compound is represented by the following chemical structure:
/>
/>
/>
/>
/>
wherein:
Z 1 an R group of a CLM as described in any aspect or embodiment herein modified to be covalently linked to L, such group selected from a bond, -C (=o) -, -CONR '-, -O-, -NR' -, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is selected from bond, H, O, OH, N, NH, NH 2 -Cl, -F, -Br, -I, methyl, optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl), optionally substituted linear or branched alkoxy (e.g., optionally substituted linear or branched C1-C6 alkoxy), -alkyl-aryl (e.g., an-alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxyl; and is also provided with
Q 1 、Q 2 、Q 3 、Q 4 、Q 5 、R 4 、L、R 1 、R 2 、R 4 、X 2 、X 4 As defined in any aspect or embodiment described herein.
In any aspect or embodiment described herein, the heterobifunctional compound is represented by the following chemical structure:
/>
/>
/>
/>
wherein:
Z 1 an R group of a CLM as described in any aspect or embodiment herein modified to be covalently linked to L, such group selected from a bond, -C (=o) -, -CONR '-, -O-, -NR' -, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is selected from bond, H, O, OH, N, NH, NH 2 Cl, -F, -Br, -I, methyl, optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl), optionally substituted linear or branched alkoxy (e.g., optionally substituted linear or branched C1-C6 alkoxy), -alkyl-aryl (e.g., optionally substituted linear or branched C1-C6 alkoxy)-alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxyl; and is also provided with
Q 1 、Q 2 、Q 3 、Q 4 、Q 5 、R 4 、L、R 1 、R 2 、X 2 、X 4As defined in any aspect or embodiment described herein.
In any aspect or embodiment described herein, the heterobifunctional compound is represented by the following chemical structure:
/>
/>
/>
Wherein:
Z 1 an R group of a CLM as described in any aspect or embodiment herein modified to be covalently linked to L, such group selected from a bond, -C (=o) -, -CONR '-, -O-, -NR' -, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is selected from bond, H, O, OH, N, NH, NH 2 Cl, -F, -Br, -I, methyl, optionally substituted straight or branched alkyl (e.g., optionally substituted)Linear or branched C1-C6 alkyl), optionally substituted linear or branched alkoxy (e.g., optionally substituted linear or branched C1-C6 alkoxy), -alkyl-aryl (e.g., an-alkyl-aryl group comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxyl; and is also provided with
Q 1 、Q 2 、Q 3 、Q 4 、Q 5 、R 4 、L、R 1a 、R 1b 、R 1c 、R 2 、X 2 、X 4 Andas defined in any aspect or embodiment described herein.
In any aspect or embodiment described herein, the heterobifunctional compound is represented by formula IIIa, formula IIIb, formula VIa, or formula VIb:
/>
wherein:
n is 0 or 1;
r is H, OH, -Cl, -F or Br;
Z 1 is nitrogen or carbon shared with the cyclic group of L;
X 4 is CH or N (preferably N);
R 1 Is that(preferably->) Wherein the dotted line is the point of attachment to the oxygen of the PTM;
R 2 is H or halogen;
the method comprises the following steps:
(preferably->
) Wherein:
R 3 is H or a linear or branched C1-C3 alkyl (e.g., methyl or ethyl);
R 3a is H, halogen, or straight or branched C1-C3 alkyl (e.g., methyl);
representation->A point of attachment to a 6 membered heteroaryl group of the PTM; and is also provided with
Represents the junction point of PTM with L or ULM, and when not present,/I>May be attached to L through an atom of the cyclic group.
L is represented by the following chemical structure:
(preferably->
),
Wherein:
W L1 is a 6 membered aromatic ring having 0, 1, 2 or 3 heteroatoms (preferably 0, 1 or 2 heteroatoms) selected from O and N (preferably N);
W L2 is a 6 membered aromatic ring having 0, 1, 2 or 3 heteroatoms (preferably 0, 1 or 2 heteroatoms) selected from O and N (preferably N);
m and n are integers independently selected from 0, 1, 2 or 3 (preferably 1); and is also provided with
The L is optionally substituted with 0, 1, 2 or 3 (preferably 0 or 1) groups selected from: -Cl, -F and C 1-3 Alkyl (e.g., methyl or ethyl).
One aspect of the present disclosure relates to a heterobifunctional compound having the chemical structure:
PTM―L―CLM,
or a pharmaceutically acceptable salt or solvate thereof,
Wherein:
(a) The CLM is a small molecule E3 ubiquitin ligase binding moiety that binds cereblon E3 ubiquitin ligase and is represented by the following chemical structure:
wherein:
w is CH 2 、C=O、SO 2 Or NH;
each X is independently selected from the group consisting of absent, O and S;
z is absent, O or S;
g is H or unsubstituted or substituted straight or branched alkyl;
each Q 1 、Q 2 、Q 3 And Q 4 N, CH or CR independently;
a is H or unsubstituted or substituted straight or branched alkyl;
n is an integer from 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched alkyl, optionally substituted alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L; and is also provided with
R 'and R' are independently selected from the group consisting of a bond, H, and optionally substituted alkyl;
represents a single bond or a double bond; and is also provided with
Represents a bond that may be stereotactic ((R) or (S)) or non-stereotactic;
(b) The PTM is a small molecule leucine rich repeat kinase 2 (LRRK 2) targeting moiety that binds to LRRK2 or a mutant form thereof, represented by the following chemical structure:
/>
wherein:
R 1 is isopropyl, tert-butyl,Wherein->Is the point of attachment to the oxygen atom of PTM;
R 2 Is hydrogen, F, cl, OH, C1-C3 alkyl or C1-C3 fluoroalkyl;
X 6 and X 7 Each independently is CH or N;
X 1 、X 2 、X 3 、X 4 and X 5 Each independently is N or CH, when CH, optionally R 2 Substitution;
X 8 is CH or N;
is an optionally substituted 3-10 membered cycloalkyl, heterocycloalkyl, bicycloalkyl, spirocycloalkyl or spiroheterocycloalkyl (containing 1-4 (e.g., 1, 2, 3 or 4) heteroatoms selected from N, O and S) (e.g., optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents); and is also provided with
The PTM isRepresents a point of attachment to the L; and is also provided with
(c) The L is a chemical linker group that covalently couples the CLM to the PTM.
One aspect of the present disclosure relates to a heterobifunctional compound having the chemical structure:
PTM―L―CLM,
or a pharmaceutically acceptable salt or solvate thereof,
wherein:
(a) The CLM is a small molecule E3 ubiquitin ligase binding moiety that binds cereblon E3 ubiquitin ligase and is represented by the following chemical structure:
wherein:
w is CH 2 、C=O、SO 2 Or NH;
each X is independently selected from the group consisting of absent, O and S;
z is absent, O or S;
g is H or unsubstituted or substituted straight or branched alkyl;
each Q 1 、Q 2 、Q 3 And Q 4 N, CH or CR independently;
A is H or unsubstituted or substituted straight or branched alkyl;
n is an integer from 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched alkyl, optionally substituted alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L; and is also provided with
R 'and R' are independently selected from the group consisting of a bond, H, and optionally substituted alkyl;
represents a single bond or a double bond; and is also provided with
Represents a bond that may be stereotactic ((R) or (S)) or non-stereotactic;
(b) The PTM is a small molecule leucine rich repeat kinase 2 (LRRK 2) targeting moiety that binds to LRRK2 or a mutant form thereof, represented by the following chemical structure:
/>
wherein:
R 1 is isopropyl, tert-butyl,Wherein->Is the point of attachment to the oxygen atom of PTM;
R 2 is hydrogen, F, cl, OH, C1-C3 alkyl or C1-C3 fluoroalkyl;
X 6 and X 7 Each independently is CH or N;
X 1 、X 2 、X 3 、X 4 and X 5 Each independently is N or CH, when CH, optionally R 2 Substitution;
X 8 is CH or N;
is an optionally substituted 3-10 membered cycloalkyl, heterocycloalkyl, bicycloalkyl, spirocycloalkyl or spiroheterocycloalkyl (containing 1-4 (e.g., 1, 2, 3 or 4) heteroatoms selected from N, O and S) (e.g., optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents); and is also provided with
The PTM isRepresents a point of attachment to the L; and is also provided with
(c) The L is a bond or a chemical linker group that covalently couples the CLM to the PTM.
In any aspect or embodiment described herein, the compound is represented by a chemical structure selected from the group consisting of:
/>
wherein:
X 2 c, CH or N;
Z 1 is a bond, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L.
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
In any aspect or embodiment described herein, the compound is represented by a chemical structure selected from the group consisting of:
wherein:
X 2 c, CH or N;
Z 1 is a bond, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L.
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
In any aspect or embodiment described herein, the compound is represented by the following chemical structure:
/>
in any aspect or embodiment described herein, the compound is represented by the following chemical structure:
In any aspect or embodiment described herein, one or more of the following:
(a) The CLM is represented by:
/>
/>
wherein:
said ULMRepresentation and placeThe connection point of the L; and is also provided with
N is a nitrogen atom shared with the L;
(b) The PTM is represented by:
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein the PTM is covalently linked to the L through an atom of the heterocycloalkyl a or a substituent thereof;
(c) The L is a linker group (L) selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Representing a point of attachment to said CLM or said PTM; or (b)
(d) A combination thereof.
In any aspect or embodiment described herein, the compound has the following chemical structure:
wherein:
X 2 c, CH or N;
Z 1 is a bond, a carbon shared with a cyclic group of L, or a nitrogen shared with a cyclic group of L.
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
In any aspect or embodiment described herein, the compound is represented by the following chemical structure:
in any aspect or embodiment described herein, the compound is represented by the following chemical structure:
In any aspect or embodiment described herein, one or more of the following:
(a) The CLM is represented by:
/>
/>
/>
wherein:
said ULMRepresents a point of attachment to the L;
c is a carbon atom shared with the L; and is also provided with
N is a nitrogen atom shared with the L;
(b) The PTM is represented by:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein the PTM is covalently linked to the L through an atom of the heterocycloalkyl a or a substituent thereof;
(c) The L is a linker group (L) selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
n is a nitrogen atom covalently linked or shared with said CLM or said PTM;
c is a carbon atom covalently linked or shared with said CLM or said PTM; and is also provided with
Representing a point of attachment to said CLM or said PTM; or (b)
(d) A combination thereof.
In any aspect or embodiment described herein, one or more of the following: the PTM is a PTM selected from the compounds of table 1, the CLM is a CLM selected from the compounds of table 1, and the L is L selected from the compounds of table 1.
In any aspect or embodiment described herein, one or more of the following: the PTM is a PTM selected from the group consisting of compounds 52-288, the CLM is a CLM selected from the group consisting of compounds 52-288, and the L is an L selected from the group consisting of compounds 52-288.
In any aspect or embodiment described herein, the compound is represented by the following chemical structure: the compounds are represented by the following chemical structures:
wherein:
n is 0 or 1;
r is H, OH, -Cl, -F or Br;
Z 1 is nitrogen or carbon shared with the cyclic group of L;
X 2 is C or N;
X 4 is CH or N (preferably N);
R 1 is that(preferably->) Wherein->Is the point of attachment to the oxygen of the PTM;
R 2 is H, -Cl or-F;
the method comprises the following steps: />
(preferably->
) Wherein:
R 3 is H, methyl or ethyl;
R 4 is H, methyl or ethyl;
representing said->A connection point with the PTM; and is also provided with
Represents the junction of said PTM with said L or said CLM, and when +.>In the absence of said->May be attached to the L through an atom of the cyclic group (e.g., carbon or nitrogen);
l is represented by the following chemical structure:
/>
wherein:
m and n are integers independently selected from 0, 1, 2 or 3 (preferably 1); and is also provided with
The L is optionally substituted with 0, 1, 2 or 3 (preferably 0 or 1) groups selected from: -Cl, -F and C 1-3 Alkyl (e.g., methyl or ethyl).
In any aspect or embodiment described herein, one or more of the following:
g is H or unsubstituted or substituted straight-chain or branched C 1-6 An alkyl group;
A is H or unsubstituted or substituted straight-chain or branched C 1-6 An alkyl group;
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched C 1-6 Alkyl, optionally substituted C 1-6 Alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L;
r 'and R' are independently selected from the group consisting of bond, H and optionally substituted C 1-6 An alkyl group; and is also provided with
Is an optionally substituted 3-10 membered cycloalkyl, an optionally substituted 3-10 membered heterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered bicycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered spirocycloalkyl, or an optionally substituted 3-10 membered spiroheterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, wherein said heteroatoms are independently selected from N, O and S.
Another aspect of the present disclosure relates to a composition comprising an effective amount of a bifunctional compound of the present disclosure and a pharmaceutically acceptable carrier.
In any aspect or embodiment described herein, the composition further comprises an additional bioactive agent.
In any aspect or embodiment described herein, the additional bioactive agent is an anti-inflammatory agent, a chemotherapeutic agent, or an immunomodulatory agent.
Another aspect of the present disclosure relates to a composition comprising a pharmaceutically acceptable carrier and an effective amount of at least one compound of the present disclosure for treating a disease, disorder, or symptom of causal relationship with LRRK2 in a subject, wherein the composition is effective to treat or ameliorate the disease, disorder, or at least one symptom of the disease or disorder.
One aspect of the present disclosure relates to a method for treating a disease, disorder, or symptom having a causal relationship with LRRK2, wherein the method comprises administering to a subject in need thereof a composition comprising a pharmaceutically acceptable carrier and an effective amount of at least one compound of the present disclosure, wherein the composition is effective to treat or ameliorate the disease, disorder, or at least one symptom of the disease or disorder. In any aspect or embodiment described herein,
another aspect of the present disclosure relates to a method of treating or preventing a disease, disorder, or condition associated with LRRK2, the method comprising providing to a patient in need thereof an effective amount of a compound as described herein or a composition comprising the compound and administering to the patient an effective amount of a compound as described herein or a composition comprising the compound, wherein the compound or composition is effective to treat or ameliorate the disease, disorder, or at least one symptom of the disease or disorder.
In any aspect or embodiment described herein, the disease or disorder is idiopathic Parkinson's Disease (PD), LRRK2 mutation-related PD, primary tauopathy, lewy body dementia, crohn's disease, leprosy, neuroinflammation, progressive supranuclear palsy, pick's disease, ftdttau, TDP-43 frontotemporal dementia, TDP-43ALS, c9orf ALS, huntington's disease, spinocerebellar ataxia (SCAs) 1, 2, 3, 6, 7 and 17, dentate nuclear pallidoluffing (DRPLA) or kennedy's disease.
The term "alkyl" in this context shall mean a straight, branched or cyclic fully saturated hydrocarbon radical, preferably C 1 -C 10 Alkyl, preferably C 1 -C 6 Alkyl, or more preferably C 1 -C 3 Alkyl groups, which may be optionally substituted with any suitable functional group or groups. Examples of alkyl groups are methyl, ethyl, n-butyl, sec-butyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, isopropyl, 2-methylpropyl, cyclopropyl, cyclopropylmethyl, cyclobutyl, cyclopentyl, cyclopentylethyl, cyclohexylethyl, cyclohexyl and the like. In certain embodiments, the alkyl groups are capped with a halogen group (At, br, cl, F or I).
The term "alkenyl" refers to a straight, branched or cyclic C containing at least one c=c bond 2 -C 10 (preferably C 2 -C 6 ) A hydrocarbon group.
The term "alkynyl" refers to a straight, branched or cyclic C containing at least one C.ident.C bond 2 -C 10 (preferably C 2 -C 6 ) A hydrocarbon group.
The term "alkylene" when used means- (CH) 2 ) n -a group (n is typically an integer from 0 to 6), which may be optionally substituted. Alkylene groups, when substituted, are preferably C on one or more methylene groups 1 -C 6 Alkyl (including cyclopropyl or tert-butyl) substituted, but may also be substituted by one or more halo groups, preferably 1 to 3 halo groups or one or two hydroxy groups, O- (C) 1 -C 6 Alkyl) groups or amino acid side chain substitutions as otherwise disclosed herein. In certain embodiments, the alkylene group may be substituted with a urethane or alkoxy group (or other suitable functional group) that may be further substituted with a polyethylene glycol chain (polyethylene glycol chain of 1 to 10, preferably 1 to 6, or more preferably 1 to 4 ethylene glycol units), the polyethylene glycol chain (preferably but not limited to at the distal end of the polyethylene glycol chain) being substituted with an alkyl chain substituted with a single halogen group (preferably a chlorine group). In still other embodiments, the alkylene (e.g., methylene) group may be substituted with a side chain group of an amino acid Group substitutions, such as natural or unnatural amino acid side chain groups, such as alanine, beta-alanine, arginine, asparagine, aspartic acid, cysteine, cystine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan, or tyrosine.
The term "unsubstituted" shall mean substituted with only hydrogen atoms. Comprising C 0 By carbon atom range is meant that carbon is absent and substituted with H. Thus C 0 -C 6 Carbon atom range of (2), 3, 4, 5 and 6, and for C 0 H replaces carbon.
The term "substituted" or "optionally substituted" shall mean that one or more substituents (independently up to five substituents, preferably up to three substituents, more preferably 1 or 2 substituents on a portion of a compound according to the present disclosure, and may include substituents that may themselves be further substituted) are present independently (i.e., each substituent is selected independently of the other substituent when more than one substitution occurs) at a carbon (or nitrogen) position anywhere on the molecule in the context, and include possible substituents: hydroxy, thiol, carboxyl, cyano (C.ident.N), nitro (NO 2 ) Halogen (preferably 1, 2 or 3 halogens, especially on an alkyl group, especially a methyl group such as trifluoromethyl), alkyl (preferably C 1 -C 10 More preferably C 1 -C 6 ) Aryl (in particular phenyl and substituted phenyl, such as benzyl or benzoyl), alkoxy (preferably C) 1 -C 6 Alkyl or aryl, including phenyl and substituted phenyl), thioether (preferably C 1 -C 6 Alkyl or aryl), acyl (preferably C 1 -C 6 Acyl), ester or thioester (preferably C 1 -C 6 Alkyl or aryl groups, including alkylene esters (so that they are attached to alkylene groups, other than preferably by C 1 -C 6 On alkyl or aryl substituted ester functions), halogen (preferably F or Cl), amine (including five or six membered cyclic alkylene amines, also including C) 1 -C 6 Alkylamine or C 1 -C 6 Dialkylamines in which the alkyl group may be substituted by one or two hydroxy groups) or optionally substituted-N (C) 0 -C 6 Alkyl) C (O) (O-C 1 -C 6 Alkyl) groups (which may optionally be substituted by polyethylene glycol chains further incorporating alkyl groups containing a single halogen, preferably chlorine, substituent), hydrazines, amide groups, which are preferably substituted by one or two C' s 1 -C 6 Alkyl (including optionally by one or two C' s 1 -C 6 Alkyl-substituted carboxamides), alkanols (preferably C 1 -C 6 Alkyl or aryl), or alkanoic acids (preferably C 1 -C 6 Alkyl or aryl). Substituents according to the present disclosure may include, for example, -SiR 1 R 2 R 3 A group, wherein R is 1 And R is 2 Each of (a) is as described further herein, and R 3 Is H or C 1 -C 6 Alkyl, preferably R 1 、R 2 、R 3 Together are C 1 -C 3 Alkyl (including isopropyl or tert-butyl). Each of the above groups may be directly attached to a substituted moiety, or alternatively, the substituents may be attached through an optionally substituted- (CH) 2 ) m- Or alternatively optionally substituted- (OCH) 2 ) m -、-(OCH 2 CH 2 ) m -or- (CH) 2 CH 2 O) m A group (which may be substituted with any one or more of the substituents described above) is attached to the substituted moiety (preferably in the case of an aryl or heteroaryl moiety). Alkylene- (CH) 2 ) m -or- (CH) 2 ) n The group or other chain as indicated above (such as ethylene glycol chain) may be substituted at any position on the chain. Preferred substituents on the alkylene include halogen or C 1 -C 6 (preferably C 1 -C 3 ) Alkyl groups, which may optionally be substituted with one or two hydroxyl groups, one or two ether groups (O-C 1 -C 6 A group), up to three halogen groups (preferably F), or side chains of amino acids and optionally substituted amides (preferably substituted carboxamides as described above) as otherwise described herein, or carbamate groups (typically having one or two C' s 0 -C 6 Alkyl substituents, one or more of which may be further substituted). In certain embodiments, alkylene (typically a single methylene group) is substituted with one or two optionally substituted C 1 -C 6 Alkyl, preferably C 1 -C 4 Alkyl, most commonly methyl or O-methyl, or side chain substitution of amino acids as otherwise described herein. In the present disclosure, a moiety in a molecule may optionally be substituted with up to five substituents, preferably up to three substituents. Most commonly, in the present disclosure, the substituted moiety is substituted with one or two substituents.
The term "substituted" (each substituent independent of any other substituent) shall also mean C in the context of its use 1 -C 6 Alkyl, C 1 -C 6 Alkoxy, halogen, amido, carboxamide, sulfone (including sulfonamide), keto, carboxyl, C 1 -C 6 Esters (oxo or carbonyl esters), C 1 -C 6 Ketone, carbamate-OC (O) -NR 1 R 2 or-N (R) 1 )-C(O)-O-R 1 Nitro, cyano and amine (including especially C 1 -C 6 alkylene-NR 1 R 2 Mono-or di-C 1 -C 6 Alkyl substituted amines, which may optionally be substituted with one or two hydroxy groups). Unless otherwise indicated in context, each of these groups contains between 1 and 6 carbon atoms. In certain embodiments, preferred substituents will include, for example, -NH-, -NHC (O) -, -O-, =o, - (CH) 2 ) m - (where m and n are in the context of 1, 2, 3, 4, 5 or 6), -S-, -S (O) -, SO 2 -or-NH-C (O) -NH-, - (CH) 2 ) n OH、-(CH 2 ) n SH、-(CH 2 ) n COO H、C 1 -C 6 Alkyl, - (CH) 2 ) n O-(C 1 -C 6 Alkyl) - (CH) 2 ) n C(O)-(C 1 -C 6 Alkyl) - (CH) 2 ) n OC(O)-(C 1 -C 6 Alkyl) - (CH) 2 ) n C(O)O-(C 1 -C 6 Alkyl) - (CH) 2 ) n NHC(O)-R 1 、-(CH 2 ) n C(O)-NR 1 R 2 、-(OCH 2 ) n OH、-(CH 2 O) n COOH、C 1 -C 6 Alkyl, - (OCH) 2 ) n O-(C 1 -C 6 Alkyl) - (CH) 2 O) n C(O)-(C 1 -C 6 Alkyl) - (OCH) 2 ) n NHC(O)-R 1 、-(CH 2 O) n C(O)-NR 1 R 2 、-S(O) 2 -R S 、-S(O)-R S (R S Is C 1 -C 6 Alkyl or- (CH) 2 ) m -NR 1 R 2 Group, NO 2 CN or halogen (F, cl, br, I, preferably F or Cl), depending on the context in which the substituent is used. R is R 1 And R is 2 In the context of H or C respectively 1 -C 6 Alkyl (which may optionally be substituted with one or two hydroxy groups or up to three halogen groups, preferably fluorine). The term "substituted" in the chemical context of the defined compounds and substituents used shall also mean optionally substituted aryl or heteroaryl or optionally substituted heterocyclyl as described further herein. Alkylene groups may also be substituted as otherwise disclosed herein, preferably optionally substituted C 1 -C 6 Alkyl (methyl, ethyl or hydroxymethyl or hydroxyethyl groups are preferred, thus providing a chiral centre), side chains of amino acid groups as otherwise described herein, amide or carbamate groups OC (O) -NR as described above 1 R 2 Radicals (wherein R 1 And R is 2 As further described herein), but many other groups may also be used as substituents. The various optionally substituted moieties may be substituted with 3 or more substituents, preferably no more than 3 substituents, and preferably with 1 or 2 substituents. It should be noted that where a substitution is required in a compound at a particular position of the molecule (primarily because of valence) but no substitution is indicated, then the substituent is interpreted or understood as H unless the context of the substitution indicates otherwise.
The term "aryl" or "aromatic" in this context refers to a substituted (as otherwise described herein) or unsubstituted monovalent aromatic group (e.g., a 5-16 membered ring) having a single ring (e.g., benzene, phenyl, benzyl, or 5, 6, 7, or 8 membered ring) or a fused ring (e.g., naphthyl, anthryl, phenanthryl, 10-16 membered ring, etc.), and may be bonded to a compound according to the present disclosure at any useful stable position on one or more of the rings or as otherwise indicated in the chemical structures presented. Other examples of aryl groups in this context may include heterocyclic aromatic ring systems, "heteroaryl" groups having one or more nitrogen, oxygen, or sulfur atoms in the ring (monocyclic ring) (such as imidazole, furyl, pyrrole, furyl, thiophene, thiazole, pyridine, pyrimidine, pyrazine, triazole, oxazole) or fused ring systems (such as indole, quinoline, indolizine, azaindolizine, benzofuran, etc.), and the like, which may be optionally substituted as described above. Heteroaryl groups that may be mentioned include nitrogen-containing heteroaryl groups such as pyrrole, pyridine, pyridone, pyridazine, pyrimidine, pyrazine, pyrazole, imidazole, triazole, triazine, tetrazole, indole, isoindole, indolizine, azaindolizine, purine, indazole, quinoline, dihydroquinoline, tetrahydroquinoline, isoquinoline, dihydroisoquinoline, tetrahydroisoquinoline, quinolizine, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, imidazopyridine, imidazotriazine, pyrazinopyridazine, acridine, phenanthridine, carbazole, carbazoline, pyrimidine, phenanthroline, phenanthrene, oxadiazole, benzimidazole, pyrrolopyridine, pyrrolopyrimidine and pyridopyrimidine; sulfur-containing aromatic heterocycles such as thiophene and benzothiophene; oxygen-containing aromatic heterocycles such as furan, pyran, cyclopentapyran, benzofuran, and isobenzofuran; and aromatic heterocycles containing 2 or more heteroatoms selected from nitrogen, sulfur and oxygen, such as thiazole, thiadiazole, isothiazole, benzoxazole, benzothiazole, benzothiadiazole, phenothiazine, isoxazole, furoxan (furazan), phenoxazine, pyrazolooxazole, imidazothiazole, thienofuran, furopyrrole, pyridooxazine, furopyridine, furopyrimidine, thienopyrimidine, oxazole, and the like, all of which may be optionally substituted.
The term "substituted aryl" refers to an aromatic carbocyclic group consisting of at least one aromatic ring or multiple condensed rings, wherein at least one ring is aromaticWherein one or more rings are substituted with one or more substituents. For example, an aryl group may comprise one or more substituents selected from the group consisting of: - (CH) 2 ) n OH、-(CH 2 ) n -O-(C 1 -C 6 ) Alkyl, - (CH) 2 ) n -O-(CH 2 ) n -(C 1 -C 6 ) Alkyl, - (CH) 2 ) n -C(O)(C 0 -C 6 ) Alkyl, - (CH) 2 ) n -C(O)O(C 0 -C 6 ) Alkyl, - (CH) 2 ) n -OC(O)(C 0 -C 6 ) Alkyl, amine, mono-or di- (C) 1 -C 6 Alkyl) amine (wherein the alkyl group on the amine is optionally substituted with 1 or 2 hydroxy or up to three halo (preferably F, cl) groups), OH, COOH, C 1 -C 6 Alkyl (preferably CH 3 )、CF 3 、OMe、OCF 3 、NO 2 Or a CN group (each of which may be substituted at the ortho, meta and/or para positions of the benzene ring, preferably at the para position), an optionally substituted phenyl group (which is itself preferably linked to the PTM group via a linker group, including ULM groups), and/or F, cl, OH, COOH, CH 3 、CF 3 、OMe、OCF 3 、NO 2 Or at least one of the CN groups (ortho, meta and/or para to the benzene ring, preferably para), optionally substituted naphthyl, optionally substituted heteroaryl, preferably optionally substituted isoxazole (including methyl substituted isoxazole), optionally substituted oxazole (including methyl substituted oxazole), optionally substituted thiazole (including methyl substituted thiazole), optionally substituted isothiazole (including methyl substituted isothiazole), optionally substituted pyrrole (including methyl substituted pyrrole), optionally substituted imidazole (including methylimidazole), optionally substituted benzimidazole or methoxybenzylimidazole, optionally substituted oxime oxazole or methylimidazole, optionally substituted diazole (including methyldiazole), optionally substituted triazole (including methyl substituted triazole), optionally substituted pyridine (including halo (preferably F) or methyl substituted pyridine) or oxapyridine (wherein the pyridine is linked to the phenyl group), optionally substituted furan, optionally substituted benzofuran, optionally substituted pyridine, or oxapyridine (wherein the pyridine is linked to the phenyl group), Optionally substituted dihydrobenzofurans, optionally substituted indoles, indolizines or azaindolizines (2, 3 or 4-azaindolizines), optionally substituted quinolines, and combinations thereof.
"carboxy" means a group-C (O) OR, wherein R is hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl OR substituted heteroaryl, and the meaning of these generic substituents has the same definition as the definition of the corresponding group as defined herein.
The term "heteroaryl" or "heteroaryl" may mean, but is in no way limited to, a 5-16 membered heteroaryl (e.g., a 5-, 6-, 7-, or 8-membered monocyclic or 10-16 membered heteroaryl having multiple fused rings), an optionally substituted quinoline (which may be attached to a pharmacophore or substituted on any carbon atom within the quinoline ring), an optionally substituted indole (including indoline), an optionally substituted indolizine, an optionally substituted azaindolizine (2, 3, or 4-azaindolizine), an optionally substituted benzimidazole, benzodiazole, benzofuran, an optionally substituted imidazole, an optionally substituted isoxazole, an optionally substituted oxazole (preferably methyl-substituted), an optionally substituted diazole, an optionally substituted triazole, tetrazole, an optionally substituted benzofuran, an optionally substituted thiophene, an optionally substituted thiazole (preferably methyl and/or mercapto-substituted), an optionally substituted isothiazole, an optionally substituted triazole (preferably methyl-, triisopropylsilyl-, optionally substituted- (CH) 2 ) m -O-C 1 -C 6 Alkyl or optionally substituted- (CH) 2 ) m -C(O)-O-C 1 -C 6 Alkyl-substituted 1,2, 3-triazoles), optionally substituted pyridines (2-, 3-or 4-pyridines) or groups according to the chemical structure:
wherein:
S c is CHR SS 、NR URE Or O;
R HET is H, CN, NO 2 Halo (preferably Cl or F), optionally substituted C 1 -C 6 Alkyl (preferably)Substituted by one or two hydroxy groups or up to three halo groups (e.g. CF 3 ) Optionally substituted O (C) 1 -C 6 Alkyl) (preferably substituted by one or two hydroxy groups or up to three halo groups) or optionally substituted alkynyl-C.ident.C-R a Wherein R is a Is H or C 1 -C 6 Alkyl (preferably C 1 -C 3 An alkyl group);
R SS is H, CN, NO 2 Halo (preferably F or Cl), optionally substituted C 1 -C 6 Alkyl (preferably substituted by one or two hydroxy groups or up to three halo groups), optionally substituted O- (C) 1 -C 6 Alkyl) (preferably substituted by one or two hydroxy groups or up to three halo groups) or optionally substituted-C (O) (C 1 -C 6 Alkyl) (preferably substituted with one or two hydroxy groups or up to three halo groups);
R URE is H, C 1 -C 6 Alkyl (preferably H or C 1 -C 3 Alkyl) or-C (O) (C 1 -C 6 Alkyl), each of which is optionally substituted with one or two hydroxy groups or up to three halogen (preferably fluoro groups), or optionally substituted heterocycles (e.g. piperidine, morpholine, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, piperazine, each of which is optionally substituted), and
Y C Is N or C-R YC Wherein R is YC Is H, OH, CN, NO 2 Halo (preferably Cl or F), optionally substituted C 1 -C 6 Alkyl (preferably substituted with one or two hydroxy groups or up to three halo groups (e.g., CF 3 ) Substituted), optionally substituted O (C) 1 -C 6 Alkyl) (preferably substituted by one or two hydroxy groups or up to three halo groups) or optionally substituted alkynyl-C.ident.C-R a Wherein R is a Is H or C 1 -C 6 Alkyl (preferably C 1 -C 3 Alkyl).
The terms "aralkyl" and "heteroarylalkyl" refer to groups comprising an aryl or respectively heteroaryl group as defined above and an alkyl and/or heteroalkyl and/or a carbocyclic and/or heterocycloalkyl ring system.
The term "arylalkyl" as used herein refers to an aryl group as defined above attached to an alkyl group as defined above. Arylalkyl groups will be attached to the parent moiety through the alkyl group, where the alkyl group is 1 to 6 carbon atoms. The aryl groups in the arylalkyl groups may be substituted as defined above.
The term "heterocycle" refers to a cyclic group containing at least one heteroatom, such as N, O or S, and may be aromatic (heteroaryl) or non-aromatic. Thus, heteroaryl moieties are included in the definition of heterocyclic ring, depending on the context in which they are used. Exemplary heteroaryl groups are described above.
Exemplary heterocycles include: azetidinyl, benzimidazolyl, 1, 4-benzodioxanyl, 1, 3-benzodioxazolyl, benzoxazolyl, benzothiazolyl, benzothienyl, dihydroimidazolyl, dihydropyranyl, dihydrofuranyl, dioxanyl, dioxolanyl, ethyleneurea, 1, 3-dioxolanyl, 1, 3-dioxane, 1, 4-dioxane, furanyl, homopiperidinyl, imidazolyl, imidazolinyl, imidazolidinyl, indolinyl, indolyl, isoquinolyl, isothiazolidinyl, isoxazolidinyl, and pharmaceutical compositions containing them isoxazolyl, morpholinyl, naphthyridinyl, oxazolidinyl, oxazolyl, pyridone, 2-pyrrolidone, pyridine, piperazinyl, N-methylpiperazinyl, piperidinyl, phthalimide, succinimide, pyrazinyl, pyrazolinyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, pyrrolyl, quinolinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydroquinoline, thiazolidinyl, thiazolyl, thienyl, tetrahydrothiophene, oxazolidine, oxetanyl, thiothiolanyl, thiadine (thiane), and the like.
The heterocyclic group may be optionally substituted with a member selected from the group consisting of: alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, amido, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxy, keto, thioketo, carboxyl, carboxyalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclyloxy, mercapto, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocycle, heterocyclyloxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SO-alkyl, -SO 2-substituted alkyl, -SO 2-aryl, oxo (=o), and-SO 2-heteroaryl. Such heterocyclic groups may have a single ring or multiple condensed rings. Examples of azacycles and heteroaryl groups include, but are not limited to, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, morpholino, piperidinyl, tetrahydrofuranyl, and the like, and N-alkoxy-nitrogen containing heterocycles. The term "heterocycle" also includes bicyclic groups in which any heterocycle is fused to a benzene ring or a cyclohexane ring or another heterocycle (e.g., indolyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, etc.).
The term "cycloalkyl" may mean, but is in no way limited to, a monovalent radical derived from a mono-or polycyclic alkyl or cycloalkyl group as defined herein, such as a saturated monocyclic hydrocarbon group having three to twenty carbon atoms in the ring, including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like. The term "substituted cycloalkyl" may refer to, but is in no way limited to, mono-or multicyclic alkyl groups and is substituted with one or more substituents such as amino, halogen, alkyl, substituted alkyl, carbonyloxy, carbonylmercapto, aryl, nitro, mercapto or sulfo, and these general substituents have the same meaning as the definition of the corresponding groups as defined in this legend.
"heterocycloalkyl" means a mono-or polycyclic alkyl group wherein at least one ring carbon atom of its cyclic structure is replaced by a heteroatom selected from the group consisting of N, O, S or P. "substituted heterocycloalkyl" refers to a mono-or polycyclic alkyl group wherein at least one ring carbon atom of its cyclic structure is replaced with a heteroatom selected from the group consisting of N, O, S or P, and the group contains one or more substituents selected from the group consisting of: halogen, alkyl, substituted alkyl, carbonyloxy, carbonylmercapto, aryl, nitro, mercapto or sulfo, and the meaning of these generic substituents is identical to the definition of the corresponding groups as defined in the legend.
The term "hydrocarbyl" shall mean a compound containing carbon and hydrogen, and which may be fully saturated, partially unsaturated or aromatic, and includes aryl, alkyl, alkenyl and alkynyl groups.
The term "independently" is used herein to indicate that the variables of the independent application vary independently from application to application.
The term "lower alkyl" refers to methyl, ethyl or propyl
The term "lower alkoxy" refers to methoxy, ethoxy or propoxy.
Exemplary CLM
Novel imide compounds
In one aspect, the present description provides CLMs that may be used to bind and recruit cereblon. In certain embodiments, the CLM is selected from the group consisting of the following chemical structures:
/>
wherein:
formulae (a 1) to (e) [ e.g., (a 1), (a 2), (a 3), (a 4), (b), (c), (d 1), (d 2) and (e)]W of (2) is independently selected from the group CH 2 、O、CHR、C=O、SO 2 NH, N, optionally substituted cyclopropyl, optionally substituted cyclobutyl and N-alkyl;
w of formulae (a 1) to (e) 3 Is C or N;
each X of formulas (a 1) through (e) is independently selected from the group consisting of the absence of a group, O, S and CH 2
Y of formulae (a 1) to (e) is selected from the group CH 2 -c=cr', NH, N-alkyl, N-aryl, N-heteroaryl, N-cycloalkyl, N-heterocyclyl, O and S;
each Z of formulas (a 1) through (e) is independently selected from the group consisting of the absence of a group, O, S and CH 2 Except that X and Z cannot both be CH 2 Or is absent;
g and G ' of formulae (a 1) to (e) are independently selected from the group H, optionally substituted straight or branched alkyl, OH, R ' OCOOR, R ' OCONRR, CH optionally substituted by R 2 -heterocyclyl and benzyl optionally substituted with R';
q of formulae (a 1) to (e) 1 -Q 4 Each of which independently represents N, CH or CR;
a of formulae (a 1) to (e) is selected from the group consisting of H, optionally substituted linear or branched alkyl, cycloalkyl, cl and F;
n in formulas (a 1) to (e) represents an integer of 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);
each R of formulas (a 1) to (e) independently includes, but is not limited to: bond, H, -C (=o) R ' (e.g., carboxyl), -CONR ' R ' (e.g., amide group), -OR ' (e.g., OH), -NR ' R ' (e.g., amine group), -SR ', -SO 2 R’、-SO 2 NR’R”、-CR’R”-、-CR’NR’R”-、(-CR’O) n’ R', optionally substituted heterocyclyl, optionally substituted aryl (e.g., optionally substituted C5-C7 aryl), optionally substituted alkyl-aryl (e.g., alkyl-aryl comprising at least one of optionally substituted C1-C6 alkyl, optionally substituted C5-C7 aryl, or a combination thereof), optionally substituted heteroaryl, optionally substituted alkyl (e.g., C1-C6 straight or branched alkyl optionally substituted with one or more halo, cycloalkyl (e.g., C3-C6 cycloalkyl), or aryl (e.g., C5-C7 aryl)), optionally substituted alkoxy (e.g., methoxy, ethoxy, butoxy, propoxy, pentoxy, or hexoxy, wherein the alkoxy may be substituted with one or more halo, alkyl, haloalkyl, fluoroalkyl, cycloalkyl (e.g., C3-C6 cycloalkyl), or aryl (e.g., C5-C7 aryl)), optionally substituted cycloalkyl, any Optionally substituted heterocyclyl, -P (O) (OR ') R', -P (O) R ', and-OP (O) (OR') R ', -OP (O) R', -Cl, -F, -Br, -I, -CF 3 、-CN、-NR’SO 2 NR’R”、-NR’CONR’R”、-CONR’COR”、-NR’C(=N-CN)NR’R”、-C(=N-CN)NR’R”、-NR’C(=N-CN)R”、-NR’C(=C-NO 2 )NR’R”、-SO 2 NR’COR”、-NO 2 、-CO 2 R’、-C(C=N-OR’)R”、-CR’=CR’R”、-CCR’、-S(C=O)(C=N-R’)R”、-SF 5 and-OCF 3 At least one of W, X, Y, Z, G, G ', R, R ', R ', Q 1 -Q 4 Or a is modified to covalently attach to PTM, a chemical linking group (L), ULM, CLM, or a combination thereof;
each of x, y and z of formulas (a 1) to (e) is independently 0, 1, 2, 3, 4, 5 or 6;
each R' and R "of formulas (a 1) to (e) is independently selected from the group consisting of a bond, H, optionally substituted straight or branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycle, -C (=o) a, and optionally substituted heterocyclyl;
n' of formulae (a 1) to (e) is an integer of 1 to 10 (e.g. 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10);
represents a single bond or a double bond; and is also provided with
Formulae (a 1) to (e)Representing a bond that may be stereotactic ((R) or (S)) or non-stereotactic.
In any aspect or embodiment described herein, the CLM comprises a chemical structure selected from the group consisting of:
/>
wherein:
formulae (a 1) to (e) [ e.g., (a 1), (a 2), (a 3), (a 4), (b), (c), (d 1), (d 2) and (e)]W is selected from the group CH 2 、O、CHR、C=O、SO 2 NH, N, optionally substituted cyclopropyl, optionally substituted cyclobutyl and N-alkyl;
w of formulae (a 1) to (e) 3 Selected from C or N;
each X of formulas (a 1) through (e) is independently selected from the group O, S and CH 2
Y of formulae (a 1) to (e) is selected from the group CH 2 -c=cr', NH, N-alkyl, N-aryl, N-heteroaryl, N-cycloalkyl, N-heterocyclyl, O and S;
each Z of formulas (a 1) through (e) is independently selected from the group O, S and CH 2 Except that X and Z cannot both be CH 2 Or is absent;
g and G ' of formulae (a 1) to (e) are independently selected from the group H, optionally substituted straight or branched alkyl, OH, R ' OCOOR, R ' OCONRR, CH optionally substituted by R 2 -heterocyclyl and benzyl optionally substituted with R';
q of formulae (a 1) to (e) 1 -Q 4 Each of which independently represents N, CH or CR;
a of formulae (a 1) to (e) is selected from the group consisting of H, optionally substituted linear or branched alkyl, cycloalkyl, cl and F;
n in formulas (a 1) to (e) represents an integer of 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);
each R of formulas (a 1) to (e) independently includes, but is not limited to: bond, H, -C (=o) R ' (e.g., carboxyl), -CONR ' R ' (e.g., amide group), -OR ' (e.g., OH), -NR ' R ' (e.g., amine group), -SR ', -SO 2 R’、-SO 2 NR’R”、-CR’R”-、-CR’NR’R”-、(-CR’O) n’ R', optionally substituted aryl (e.g., optionally substituted C5-C7 aryl), optionally substituted alkyl-aryl (e.g., comprisingAn optionally substituted C1-C6 alkyl, an optionally substituted C5-C7 aryl, or an alkyl-aryl of at least one of a combination thereof), an optionally substituted heteroaryl, an optionally substituted linear or branched alkyl (e.g., a C1-C6 linear or branched alkyl optionally substituted with one or more halogens, cycloalkyl (e.g., C3-C6 cycloalkyl), or aryl (e.g., C5-C7 aryl), an optionally substituted alkoxy (e.g., methoxy, ethoxy, butoxy, propoxy, pentoxy, or hexoxy); wherein the alkoxy groups may be substituted with one OR more halo, alkyl, haloalkyl, fluoroalkyl, cycloalkyl (e.g., C3-C6 cycloalkyl), OR aryl (e.g., C5-C7 aryl) substituted), optionally substituted cycloalkyl, optionally substituted heterocyclyl, -P (O) (OR ') R', -P (O) R ', -OP (O) (OR') R ', -OP (O) R', -Cl, -F, -Br, -I, -CF 3 、-CN、-NR’SO 2 NR’R”、-NR’CONR’R”、-CONR’COR”、-NR’C(=N-CN)NR’R”、-C(=N-CN)NR’R”、-NR’C(=N-CN)R”、-NR’C(=C-NO 2 )NR’R”、-SO 2 NR’COR”、-NO 2 、-CO2R’、-C(C=N-OR’)R”、-CR’=CR’R”、-CCR’、-S(C=O)(C=N-R’)R”、-SF 5 and-OCF 3 Wherein W, X, Y, Z, G, G ', R, R', R ", Q 1 -Q 4 Or at least one of a is covalently linked (directly or indirectly, e.g., through a functional group or atom such as O, S, N) to a PTM, a chemical linking group (L), ULM, CLM, or a combination thereof;
Each of x, y and z of formulas (a 1) to (e) is independently 0, 1, 2, 3, 4, 5 or 6;
each R' and R "of formulas (a 1) to (e) is independently selected from the group consisting of a bond, H, optionally substituted straight or branched alkyl, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycle, -C (=o) a, and optionally substituted heterocyclyl;
n' of formulas (a 1) to (e) is an integer of 1 to 10 (e.g., 1 to 4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10); and is also provided with
Formulae (a) to (f)Representing a bond that may be stereotactic ((R) or (S)) or non-stereotactic.
In any aspect or embodiment described herein, the CLM or ULM is selected from the structures of formula (g):
wherein:
w of formula (g) is selected from the group CH 2 O, C = O, NH and N-alkyl;
a of formula (g) is H, methyl or optionally substituted straight or branched alkyl;
n is an integer from 1 to 4;
each R of formula (g) is independently selected from a bond, H, O, OH, N, NH, NH 2 -Cl, -F, -Br, -I, methyl, optionally substituted linear or branched alkyl (e.g., optionally substituted linear or branched C1-C6 alkyl), optionally substituted linear or branched alkoxy (e.g., optionally substituted linear or branched C1-C6 alkoxy), -alkyl-aryl (e.g., an-alkyl-aryl group comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxyl), wherein at least one R or W is modified to covalently bond to a PTM, chemical linking group (L), ULM, CLM, or a combination thereof; and is also provided with
Of formula (g)Representing a bond that may be stereotactic ((R) or (S)) or non-stereotactic.
In any aspect or embodiment described herein, the CLM or ULM is selected from the group consisting of:
wherein:
w is c=o or CH 2
N is a nitrogen atom covalently attached to the PTM or linker, or a nitrogen atom shared with the PTM or linker (L) (e.g., a heteroatom shared with an optionally substituted heterocyclyl of the linker (L) or PTM); and is also provided with
Represents the point of attachment of the CLM or ULM to the linker (L) or PTM.
In any aspect or embodiment described herein, each R is independently selected from: H. o, OH, N, NH, NH 2 A C1-C6 alkyl group, a C1-C6 alkoxy group, -an alkyl-aryl group (e.g., an-alkyl-aryl group comprising at least one of a C1-C6 alkyl group, a C4-C7 aryl group, or a combination thereof), an aryl group (e.g., a C5-C7 aryl group), an amine, an amide, or a carboxyl group.
In any aspect or embodiment described herein, at least one R (e.g., selected from the following H, O, OH, N, NH, NH 2 A C1-C6 alkyl, a C1-C6 alkoxy, -alkyl-aryl (e.g., an-alkyl-aryl comprising at least one of a C1-C6 alkyl, a C4-C7 aryl, or a combination thereof), an aryl (e.g., an R group of a C5-C7 aryl), an amine, an amide, or a carboxyl), or W is modified to covalently bond to a PTM, a chemical linker group (L), ULM, CLM, or a combination thereof.
In any aspect or embodiment described herein, W, X, Y, Z, G, G ', R, R', R ", Q1-Q4 and a of formulas (a) to (g) can be independently covalently coupled to a linker and/or a linker linking one or more PTM, ULM or CLM groups.
In any aspect or embodiment described herein, N is an integer from 1 to 4, and each R is an independently selected functional group or atom, such as O, OH, N, -Cl, -F, C1-C6 alkyl, C1-C6 alkoxy, -alkyl-aryl (e.g., an-alkyl-aryl comprising at least one of C1-C6 alkyl, C4-C7 aryl, or a combination thereof), aryl (e.g., C5-C7 aryl), amine, amide, or carboxyl on the aryl or heteroaryl of the CLM, and optionally one of them is modified to covalently bond to PTM, a chemical linker group (L), ULM, CLM, or a combination thereof.
More specifically, non-limiting examples of CLMs include those shown below as well as those "hybrid" molecules resulting from a combination of one or more of the different features shown in the following molecules, wherein at least one R or W is modified to covalently bind to a PTM, chemical linking group (L), ULM, CLM, or a combination thereof.
/>
/>
/>
/>
/>
/>
/>
In any aspect or embodiment described herein, the CLM comprises a chemical structure selected from the group consisting of:
/>
/>
/>
Wherein:
w is selected from CH 2 、O、CHR、C=O、SO 2 NH, N, optionally substituted cyclopropyl, optionally substituted cyclobutyl, and N-alkyl (e.g., CH 2 、CHR、C=O、SO 2 NH and N-alkyl);
Q 1 、Q 2 、Q 3 、Q 4 and Q 5 Each of which is independently N, CH or CR';
R 1 is a bond, H, OH, CN, C1-C3 alkyl or c=o;
R 2 is absent, H, OH, CN, C1-C3 alkyl, CHF 2 、CF 3 CHO or C (=o) NH 2
R 3 Is a bond, H, alkyl (e.g., C1-C6 or C1-C3 alkyl), substituted alkyl (e.g., substituted C1-C6 or C1-C3 alkyl), alkoxy (e.g., C1-C6 or C1-C3 alkoxy), or substituted alkoxy (e.g., substituted C1-C6 or C1-C3 alkoxy);
R 4 is a bond, H, alkyl or substituted alkyl;
R 5 and R is 6 Each independently is a bond, H, halogen, C (=O) R', CN, OH or CF 3
X is C, CH, c=o or N;
X 1 is C= O, N, CH or CH 2
Each R' is independently a bond, H, halogen, amine, alkyl (e.g., C1-C3 alkyl), substituted alkyl (e.g., substituted C1-C3 alkyl), alkoxy (e.g., C1-C3 alkoxy), substituted alkoxy (e.g., substituted C1-C3 alkoxy), NR 2 R 3 、C(=O)OR 2 Or optionally substituted phenyl;
n is an integer from 0 to 4;
is a single bond or a double bond; and is also provided with
CLM is covalently attached to PTM, chemical linker group (L), ULM, CLM, or a combination thereof.
In any aspect or embodiment described herein, the CLM is represented by the following chemical structure:
in any aspect or embodiment described herein, the CLM is formed by an R group (such as R, R 1 、R 2 、R 3 、R 4 Or R'), W, X, or a Q group (such as Q) 1 、Q 2 、Q 3 、Q 4 Or Q 5 ) Covalently bonded to a PTM or chemical linker group (L).
In any aspect or embodiment described herein, the CLM is provided by W, X, R, R 1 、R 2 、R 3 、R 4 、R 5 、R’、Q 1 、Q 2 、Q 3 、Q 4 And Q 5 Covalently bonded to a PTM or chemical linker group (L).
In any aspect or embodiment described herein, W, X, R 1 、R 2 、R 3 、R 4 、R’、Q 1 、Q 2 、Q 3 、Q 4 And Q 5 The linker may be covalently coupled independently to the linker and/or to one or more PTM, ULM or CLM groups.
More specifically, non-limiting examples of CLMs include those shown below as well as "hybrid" molecules or compounds resulting from combining one or more features of the following compounds:
/>
/>
wherein:
w is selected from the group CH 2 、CHR、C=O、SO 2 NH and N-alkyl;
R 1 is absent (i.e., bond), H, CH, CN, or C1-C3 alkyl;
R 2 is a bond, H or C1-C3 alkyl;
R 3 is a bond, H, alkyl, substituted alkyl, alkoxy or substituted alkoxy;
R 4 is a bond, methyl or ethyl;
R 5 is a bond, H or halogen;
R 6 is a bond, H or halogen;
n is an integer from 0 to 4;
Each R and R' is independently a bond, H, a functional group or atom (e.g., H, halogen (e.g., -Cl or-F), an amine, C1-C3 alkyl, C1-C3 alkoxy, NR 2 R 3 OR C (=O) OR 2 ) The method comprises the steps of carrying out a first treatment on the surface of the Or the point of attachment of PTM or chemical linker group (L),
Q 1 and Q 2 Each independently is N or C substituted with a group independently selected from H and C1-C3 alkyl; and is combined withAnd is also provided with
Is a single bond or a double bond.
In any aspect or embodiment described herein, W, R 1 、R 2 、Q 1 、Q 2 、Q 3 、Q 4 R, and R' may be independently covalently coupled to a linker and/or a linker attached to one or more PTM groups.
In any aspect or embodiment described herein, R 1 、R 2 、Q 1 、Q 2 、Q 3 、Q 4 R, and R' may be independently covalently coupled to a linker and/or a linker attached to one or more PTM groups.
In any aspect or embodiment described herein, Q 1 、Q 2 、Q 3 、Q 4 R, and R' may be independently covalently coupled to a linker and/or a linker attached to one or more PTM groups.
In any aspect or embodiment described herein, R is a bond or is modified to covalently bond to a linker group (L) or PTM or a combination thereof.
As will be apparent, in any aspect or embodiment described herein, R, R', R "of the CLM 1 、R 2 、R 3 、R 4 、R 5 And R is 6 May be a key.
In any aspect or embodiment described herein, the CLM is selected from the group consisting of:
/>
wherein R' is halogen and R 1 As described herein.
In some cases, a "CLM" may be an imide that binds to the cereblon E3 ligase. These imide and linker attachment points may be, but are not limited to, one of the following structures:
/>
in any aspect or embodiment described herein, the ULM is selected from the group consisting of:
/>
/>
/>
wherein:
ULM (ULM)Represents the point of attachment to the linker group or PTM;
n is a nitrogen atom shared with the chemical linker group or PTM;
c is a carbon atom shared with a chemical linker group or PTM, and
W、Q 4 and Q 5 Each as defined in any aspect or embodiment described herein.
Described hereinIn any aspect or embodiment, each R of the CLM is selected from H, O, OH, N, NH, NH 2 、Cl、-F、-Br、-I、CN、CF 3 Optionally substituted straight-chain or branched C 1-3 Alkyl, optionally substituted straight or branched C 1-3 Alkoxy, amide, and carboxyl.
In any aspect or embodiment described herein, each R of the CLM is selected from H, O, OH, N, NH, NH 2 、Cl、-F、-Br、-I、CN、CF 3 Methyl, methoxy, ethoxy, amide and carboxyl.
In any aspect or embodiment described herein, each R of the CLM is selected from H, O, OH, N, NH, NH 2 、Cl、-F、-Br、-I、CN、CF 3 Methyl, methoxy and ethoxy.
In any aspect or embodiment described herein, the CLM is represented by the following chemical structure:
in any aspect or embodiment described herein, the CLM has a chemical structure represented by:
in any aspect or embodiment described herein, each R is selected from H, O, OH, NH, NH 2 、-Cl、-F、CN、CF 3 Optionally substituted straight-chain or branched C 1-3 Alkyl, optionally substituted straight or branched C 1-3 An alkoxy group.
In any aspect or embodiment described herein, each R is selected from H, O, OH, NH, NH 2 、-Cl、-F、-CN、CF 3 Methyl, methoxy and ethoxy.
Exemplary Joint
In certain embodiments, the compounds as described herein comprise a moiety that is attached to the surface of the substrate by a chemical linker(L) PTM chemically linked to ULM (e.g., CLM). In certain embodiments, the linker group L comprises one or more covalently linked building blocks (e.g., -A) L 1…(A L ) q -or- (A) L ) q (-), wherein A L 1 Is a group coupled to PTM, and (A) L ) q Is a group coupled to ULM.
In any aspect or embodiment described herein, the linkage of the linker (L) to the ULM (e.g., CLM) is a stable L-ULM linkage. For example, in any aspect or embodiment described herein, when the linker (L) and ULM are connected by a heteroatom (e.g., N, O, S), any additional heteroatom (if present) is substituted with at least one carbon atom (e.g., -CH) 2 (-), such as acetal or aminal groups. As a further example, in any aspect or embodiment described herein, when the linker (L) and ULM are connected by a heteroatom, the heteroatom is not part of the ester.
In any aspect or embodiment described herein, the linker group L is a bond or is represented by formula- (a) L ) q -a chemical linker group represented, wherein a is a chemical moiety and q is an integer from 1-100 or 1-80 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80), and wherein L is covalently bonded to both PTM and ULM and binding of PTM to a protein target and ULM to an E3 ubiquitin ligase are provided to cause ubiquitination of the target protein.
In any aspect or embodiment described herein, the linker group L is a bond or is represented by formula- (a) L ) q A chemical linker group represented by (1) wherein A is a chemical moiety and q is an integer of 6-30 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22,23. 24 or 25), and wherein L is covalently bound to both PTM and ULM and sufficiently close to provide binding of PTM to a protein target and binding of ULM to E3 ubiquitin ligase to result in ubiquitination of the target protein.
In any aspect or embodiment described herein, the linker group L is- (a) L ) q -, wherein:
(A L ) q is a group that connects the ULM (e.g., CLM) to the PTM;
q of the linker is an integer greater than or equal to 1;
each A L Independently selected from the group consisting of: bond, CR L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 、C≡C、SiR L1 R L2 、P(O)R L1 、P(O)OR L1 、NR L3 C(=NCN)NR L4 、NR L3 C(=NCN)、NR L3 C(=CNO 2 )NR L4 Optionally by 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 1-6R L1 And/or R L2 Aryl substituted with groups and optionally with 1-6R L1 And/or R L2 Heteroaryl substituted with a group, wherein R L1 Or R is L2 Each independently optionally linked to other groups to form optionally substituted 1-4R L5 A cycloalkyl or heterocyclyl moiety substituted with a group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl group,NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
In certain embodiments, q is an integer greater than or equal to 1.
In any aspect or embodiment described herein, for example, when q of the linker is greater than 2, (a) L ) q Is A as L 1 And (A) L ) q Wherein the linker couples the PTM to the ULM.
Any of the parties described hereinIn a face or embodiment, for example, when q of the linker is 2, A L 2 Is connected to A L 1 And a group attached to ULM.
In any aspect or embodiment described herein, for example, when q of the linker is 1, the structure of the linker group L is-a L 1 -, and A L 1 Is a group that connects the ULM moiety to the PTM moiety.
In any aspect or embodiment described herein, unit a of linker (L) L Comprising a group represented by a general structure selected from the group consisting of:
-NR(CH 2 ) n - (lower alkyl) -, -NR (CH) 2 ) n - (lower alkoxy) -, -NR (CH) 2 ) n - (lower alkoxy) -OCH 2 -、-NR(CH 2 ) n - (lower alkoxy) - (lower alkyl) -OCH 2 -、-NR(CH 2 ) n - (cycloalkyl) - (lower alkyl) -OCH 2 -、-NR(CH 2 ) n - (heterocycloalkyl) -, NR (CH) 2 CH 2 O) n - (lower alkyl) -O-CH 2 -、-NR(CH 2 CH 2 O) n - (heterocycloalkyl) -O-CH 2 -、-NR(CH 2 CH 2 O) n -aryl-O-CH 2 -、-NR(CH 2 CH 2 O) n - (heteroaryl) -O-CH 2 -、-NR(CH 2 CH 2 O) n - (cycloalkyl) -O- (heteroaryl) -O-CH 2 -、-NR(CH 2 CH 2 O) n - (cycloalkyl) -O-aryl-O-CH 2 -、-NR(CH 2 CH 2 O) n - (lower alkyl) -NH-aryl-O-CH 2 -、-NR(CH 2 CH 2 O) n - (lower alkyl) -O-aryl-CH 2 、-NR(CH 2 CH 2 O) n -cycloalkyl-O-aryl-, -NR (CH) 2 CH 2 O) n -cycloalkyl-O- (heteroaryl) -, -NR (CH) 2 CH 2 ) n - (cycloalkyl) -O- (heterocyclyl) -CH 2 、-NR(CH 2 CH 2 ) n - (heterocyclyl) -CH 2 and-N (R1R 2) - (heterocyclyl) -CH 2 The method comprises the steps of carrying out a first treatment on the surface of the Wherein the method comprises the steps of
N of the linker may be 0 to 10;
r of the linker may be H or lower alkyl; and is also provided with
R1 and R2 of the linker may form a ring with the linker N.
In any aspect or embodiment described herein, the linker (L) comprises optionally substituted C 1 -C 50 Alkyl (e.g., C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 7 、C 8 、C 9 、C 10 、C 11 、C 12 、C 13 、C 14 、C 15 、C 16 、C 17 、C 18 、C 19 、C 20 、C 21 、C 22 、C 23 、C 24 、C 25 、C 26 、C 27 、C 28 、C 29 、C 30 、C 31 、C 32 、C 33 、C 34 、C 35 、C 36 、C 37 、C 38 、C 39 、C 40 、C 41 、C 42 、C 43 、C 44 、C 45 、C 46 、C 47 、C 48 、C 49 Or C 50 Alkyl, and includes all implied subranges, e.g., C1-C10, C1-C20; C2-C10, C2-20; C10-C20, C10-C50, etc.), wherein each carbon is optionally substituted or replaced independently with: (1) a heteroatom selected from N, O, S, P or Si atoms having the appropriate number of hydrogens, substitutions, or both to satisfy valence, (2) optionally substituted cycloalkyl or bicyclocycloalkyl, (3) optionally substituted heterocycloalkyl or bicycloheterocycloalkyl, (4) optionally substituted aryl or bicycloaryl, or (5) optionally substituted heteroaryl or bicycloheteroaryl. In any aspect or embodiment described herein, the linker (L) has no heteroatom-heteroatom linkage (e.g., no heteroatom covalent linkage or adjacent positioning).
In any aspect or embodiment described herein, the linker (L) comprises optionally substituted C 1 -C 50 Alkyl (e.g., C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 7 、C 8 、C 9 、C 10 、C 11 、C 12 、C 13 、C 14 、C 15 、C 16 、C 17 、C 18 、C 19 、C 20 、C 21 、C 22 、C 23 、C 24 、C 25 、C 26 、C 27 、C 28 、C 29 、C 30 、C 31 、C 32 、C 33 、C 34 、C 35 、C 36 、C 37 、C 38 、C 39 、C 40 、C 41 、C 42 、C 43 、C 44 、C 45 、C 46 、C 47 、C 48 、C 49 Or C 50 Alkyl), wherein:
each carbon is optionally replaced by: CR (computed radiography) L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 、C≡C、SiR L1 R L2 、P(O)R L1 、P(O)OR L1 、NR L3 C(=NCN)NR L4 、NR L3 C(=NCN)、NR L3 C(=CNO 2 )NR L4 Optionally by 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 1-6R L1 And/or R L2 Aryl substituted by radicals or optionally substituted by 0-6R L1 And/or R L2 Heteroaryl substituted with a group, wherein R L1 Or R is L2 Each independently optionally linked to other groups to form optionallyIs covered by 1-4R L5 A cycloalkyl or heterocyclyl moiety substituted with a group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
In any aspect or embodiment described herein, the linker group is optionally takenSubstituted optionally substituted C 1 -C 50 Alkyl (e.g., C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 7 、C 8 、C 9 、C 10 、C 11 、C 12 、C 13 、C 14 、C 15 、C 16 、C 17 、C 18 、C 19 、C 20 、C 21 、C 22 、C 23 、C 24 、C 25 、C 26 、C 27 、C 28 、C 29 、C 30 、C 31 、C 32 、C 33 、C 34 、C 35 、C 36 、C 37 、C 38 、C 39 、C 40 、C 41 、C 42 、C 43 、C 44 、C 45 、C 46 、C 47 、C 48 、C 49 Or C 50 Alkyl, and includes all implied subranges such as C1-C10, C1-C20; C2-C10, C2-20; C10-C20, C10-C50, etc.), wherein each carbon atom is optionally substituted or replaced with: o, N, S, P or Si atoms with an appropriate number of hydrogens, substitutions (e.g., OH, halo, alkyl, methyl, ethyl, haloalkyl, hydroxyalkyl, alkoxy, methoxy, etc.), or both to satisfy valence; optionally substituted aryl (e.g., optionally substituted C5 or C6 aryl) or bicyclic aryl (e.g., optionally substituted C5-C20 bicyclic heteroaryl); optionally substituted heteroaryl (e.g., optionally substituted C5 or C6 heteroaryl) or bicyclic heteroaryl (e.g., optionally substituted heteroaryl or bicyclic heteroaryl having one or more heteroatoms selected from N, O, S, P and Si with an appropriate amount of hydrogen, substitution (e.g., OH, halo, alkyl, methyl, ethyl, haloalkyl, hydroxyalkyl, alkoxy, methoxy, etc.), or both to satisfy valence); optionally substituted C1-C6 alkyl; optionally substituted C1-C6 alkenyl; optionally substituted C1-C6 alkynyl; optionally substituted cycloalkyl (e.g., optionally substituted C3-C7 cycloalkyl) or bicyclocycloalkyl (e.g., optionally substituted C5-C20 bicyclocycloalkyl); or optionally substituted heterocycloalkyl (e.g., optionally Substituted 3-, 4-, 5-, 6-, or 7-membered heterocyclic groups) or bicyclic heteroalkyl (e.g., optionally substituted heterocycloalkyl bicyclic heteroalkyl having one or more heteroatoms selected from N, O, S, P or Si atoms with appropriate amounts of hydrogen, substitution (e.g., OH, halo, alkyl, methyl, ethyl, haloalkyl, hydroxyalkyl, alkoxy, methoxy, etc.), or both to satisfy valence). In any aspect or embodiment described herein, the optionally substituted alkyl linker is optionally substituted with one or more OH, halo, linear or branched C1-C6 alkyl (such as methyl or ethyl), linear or branched C1-C6 haloalkyl, linear or branched C1-C6 hydroxyalkyl, or linear or branched C1-C6 alkoxy (e.g., methoxy).
In any aspect or embodiment described herein, the linker (L) has no heteroatom-heteroatom linkage (e.g., no heteroatom covalent linkage or adjacent positioning).
In any aspect or embodiment described herein, the linker (L) comprises from about 1 to about 50 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50) optionally substituted alkylene glycol units, wherein carbon or oxygen may be substituted with a heteroatom selected from N, S, P or Si atoms having an appropriate number of hydrogens to satisfy the valence.
In any aspect or embodiment described herein, unit a of linker (L) L Comprising a structure selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein N is a nitrogen atom covalently linked or shared with said ULM or said PTM, C is a carbon atom covalently linked or shared with said ULM or said PTM, andrepresenting the connection point with the CLM or the PTM.
In any aspect or embodiment described herein, unit a of linker (L) L Comprising a structure selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
the linker is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen and methyl (preferably independently selected from halogen);
c is a carbon atom covalently linked or shared with said CLM or said PTM;
n is a nitrogen atom covalently linked or shared with the ULM or the PTM; and is also provided with
Each m, n, o, p, q and r is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 (preferably independently 0, 1, 2 or 3).
In any aspect or embodiment described herein, unit a of linker (L) L Selected from:
/>
/>
/>
/>
wherein N is a nitrogen atom covalently linked to or shared with said ULM or PTM.
In any aspect or embodiment described herein, unit a of linker (L) L Comprising a group represented by a general structure selected from the group consisting of:
/>
and is also provided with
Wherein,
m, n, o, p, q and r of the linker are independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20;
when m, N, O, p, q and r are zero, no N-O or O-O bond is present,
x of the linker is H or F;
/>
/>
/>
/>
wherein each n and m of the linker may independently be 0, 1, 2, 3, 4, 5 or 6.
In any aspect or embodiment described herein, unit a of linker (L) L Selected from the group consisting of:
/>
wherein each m and n is independently selected from 0, 1, 2, 3, 4, 5 or 6.
In any aspect or embodiment described herein, unit a of linker (L) L Selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein each m, n, o, p, q, r and s is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.
In any aspect or embodiment described herein, unit a of linker (L) L Selected from the group consisting of:
/>
/>
/>
/>
in any aspect or embodiment described herein, the linker (L) comprises a structure selected from the structures shown below:
Wherein:
W L1 and W is L2 Each independently is a non-existent 4-8 membered ring having 0-4 heteroatoms, optionally substituted with R Q Substituted, each R Q Independently H, halo, OH, CN, CF 3 Optionally substituted straight-chain or branched C 1-6 Alkyl, optionally substituted straight or branched C 1-6 Alkoxy, or 2R Q The groups together with the atoms to which they are attached form a 4-8 membered ring system containing 0-4 heteroatoms;
each Y L1 Independently a bond; optionally substituted straight or branched C 1-6 An alkoxy group; optionally substituted straight or branched C 1-6 Alkyl, and one or more C atoms are optionally replaced by O or NR YL1 Replacement; optionally substituted C 2-6 Olefins, with one or more C atoms optionally replaced by O; or optionally substituted C 2-6 Alkynes, with one or more C atoms optionally replaced by O;
each R YL1 Is H or optionally substituted straight-chain or branched C 1-6 An alkyl group;
n is an integer from 0 to 10; and is also provided with
And->Representing the point of attachment to the PTM or ULM.
In any aspect or embodiment described herein, the linker (L) comprises a structure selected from the structures shown below:
wherein:
W L1 and W is L2 Each independently is absent, piperazine, piperidine, morpholine, optionally substituted with R Q Substituted, each R Q Independently H, -Cl-, -F-, OH, CN, CF 3 Optionally substituted straight-chain or branched C 1-6 Alkyl (e.g. methyl, ethyl), optionally substituted straight or branched C 1-6 Alkoxy (e.g., methoxy, ethoxy);
each Y L1 Independently a bond; optionally substituted straight or branched C 1 -C 6 An alkoxy group; optionally substituted straight or branched C 1-6 Alkyl, and one or more C atoms are optionally replaced by O or NR YL1 Replacement; optionally substituted C 2-6 Olefins, with one or more C atoms optionally replaced by O; optionally substituted C 2-6 Alkynes, with one or more C atoms optionally replaced by O;
each R YL1 Is H or optionally substituted straight-chain or branched C 1-6 Alkyl (e.g., methyl, ethyl);
n is an integer from 0 to 10; and is also provided with
And->Representing the point of attachment to the PTM or ULM.
In any aspect or embodiment described herein, the linker (L) comprises a structure selected from the structures shown below:
wherein:
W L1 and W is L2 Each independently is absent; an aryl group; heteroaryl; a ring; a heterocycle; c (C) 1-6 Alkyl, and one or more C atoms are optionally replaced by O or NR YL1 Replacement; c (C) 2-6 Olefins, with one or more C atoms optionally replaced by O; c (C) 2-6 Alkynes, with one or more C atoms optionally replaced by O; a double ring; biaryl; a diheteroaryl group; or a bisheterocycle, each optionally substituted with RQ, each RQ is independently H, halogen, OH, CN, CF 3 Hydroxyl, nitro, C.ident.CH, C 2-6 Alkenyl, C 2-6 Alkynyl, optionally substituted straight or branched C 1 -C 6 Alkyl, optionally substituted straight or branched C 1-6 Alkoxy, optionally substituted OC 1-3 Alkyl (e.g., optionally substituted with 1 or more-F), OH, NH 2 、NR Y1 R Y2 CN, or 2 RQ groups together with the atoms to which they are attached form a 4-8 membered ring system containing 0-4 heteroatoms;
each Y L1 Independently a bond; NR (NR) YL1 ;O;S;NR YL2 ;CR YL1 R YL2 ;C=O;C=S;SO;SO 2 The method comprises the steps of carrying out a first treatment on the surface of the Optionally substituted straight or branched C 1 -C 6 An alkoxy group; or optionally substituted straight-chain or branched C 1 -C 6 Alkyl, and one or more C atoms are optionally replaced with O;
Q L is an optionally bridged 3-to 6-membered alicyclic, bicyclic or aromatic ring having 0-4 heteroatoms, optionally substituted with 0-6R Q Substituted, each R Q Independently H, optionally substituted straight or branched C 1-6 Alkyl (e.g. optionally substituted with 1 or more halogens or C 1-6 Alkoxy substitution), or 2R Q The groups together with the atoms to which they are attached form a 3-8 membered ring system containing 0-2 heteroatoms;
each R YL1 And R is YL2 Independently H, OH, optionally substituted straight or branched C 1-6 Alkyl (e.g. optionally substituted with 1 or more halogens or C 1-6 Alkoxy substitution), or R YL1 And R is YL2 Together with the atoms to which they are attached, form a 3-8 membered ring system containing 0-2 heteroatoms;
n is an integer from 0 to 10; and is also provided with
And->Representing the point of attachment to the PTM or ULM.
In any aspect or embodiment described herein, the PTM, ULM, or L is a chemical moiety as described or exemplified herein. That is, any of the PTMs, ULMs, or ls described may be combined with any other PTMs, ULMs, or ls as described herein in any combination.
In any aspect or embodiment described herein, the linker (L) comprises a structure selected from the structures shown below:
wherein:
W L1 and W is L2 Each independently is absent; cyclohexane; cyclopentane; piperazine; piperidine; morpholine; c (C) 1-6 Alkyl, and one or more C atoms are optionally replaced by O or NR YL1 Replacement; c (C) 2-6 Olefins, with one or more C atoms optionally replaced by O; c (C) 2-6 olefins And one is provided withOne or more C atoms are optionally replaced by O; or C 2-6 Alkynes, with one or more C atoms optionally replaced by O, each optionally substituted with RQ, each R Q Independently H, -Cl, -F, OH, CN, CF3, hydroxy, optionally substituted straight or branched C 1-6 Alkyl (e.g., methyl, ethyl), or optionally substituted straight or branched C 1-6 An alkoxy group;
each Y L1 Independently a bond; NR (NR) YL1 ;O;CR YL1 R YL2 The method comprises the steps of carrying out a first treatment on the surface of the C=o; optionally substituted straight or branched C 1 -C 6 An alkoxy group; or optionally substituted straight-chain or branched C 1-6 Alkyl, and one or more C atoms are optionally replaced by O or NR YL1 Replacement; c (C) 2-6 Olefins, with one or more C atoms optionally replaced by O; c (C) 2-6 Alkynes, with one or more C atoms optionally replaced by O;
Q L is a 3-6 membered heterocyclic, heterobicyclic or heteroaryl ring, optionally substituted with 0-6R Q Substituted, each R Q Independently H or optionally substituted straight or branched C 1-6 Alkyl (e.g. optionally substituted with 1 or more halo, C 1-6 Alkoxy substitution);
each R YL1 And R is YL2 Independently H or optionally substituted straight or branched C 1-6 Alkyl (e.g., methyl, ethyl, optionally substituted with 1 or more halogens or C 1-6 Alkoxy substitution);
n is an integer from 0 to 10; and is also provided with
And->Representing the point of attachment to the PTM or ULM.
In any aspect or embodiment described herein, the L comprises a chemical structural unit represented by the formula: - (A) L ) q -,
Wherein:
-(A L ) q -is a group attached to the CLM and the PTM;
q is an integer greater than or equal to 1;
each A L Independently selected from CR L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 C.ident.C, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 0-6R L1 And/or R L2 Aryl substituted with groups and optionally with 1-6R L1 And/or R L2 A group-substituted heteroaryl group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
In any aspect or embodiment described herein, the L comprises optionally substituted C 1 -C 50 Alkyl (e.g., C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 7 、C 8 、C 9 、C 10 、C 11 、C 12 、C 13 、C 14 、C 15 、C 16 、C 17 、C 18 、C 19 、C 20 、C 21 、C 22 、C 23 、C 24 、C 25 、C 26 、C 27 、C 28 、C 29 、C 30 、C 31 、C 32 、C 33 、C 34 、C 35 、C 36 、C 37 、C 38 、C 39 、C 40 、C 41 、C 42 、C 43 、C 44 、C 45 、C 46 、C 47 、C 48 、C 49 Or C 50 Alkyl), wherein:
each carbon is optionally replaced by: CR (computed radiography) L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 C.ident.C, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 1-6R L1 And/or R L2 Aryl substituted by radicals or optionally substituted by 1-6R L1 And/or R L2 A group-substituted heteroaryl group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
In any aspect or embodiment described herein, the L is selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
Wherein:
the chemical linker group is optionally substituted with halogen;
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Each m, n, o, p, q and r of the L is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20.
In any aspect or embodiment described herein, the L is selected from the group consisting of:
/>
wherein:
the chemical linker group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen and methyl (preferably independently selected from halogen);
c is a carbon atom covalently linked or shared with said CLM or said PTM;
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Each m, n, and o of the L is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.
Exemplary PTM
In one aspect of the disclosure, a PTM group (also referred to as an LTM group) binds to the target protein LRRK2 or a mutant form thereof.
The compositions described below exemplify members of the LRRK2 binding moiety that can be used according to the invention. These binding moieties are preferably linked to ubiquitin ligase binding moiety (CLM) by a chemical linking group so that LRRK2 protein (which binds to LTM) is presented in proximity to ubiquitin ligase for ubiquitination and subsequent degradation.
In some cases, the term "target protein" is used to refer to LRRK2 protein, which is a member of the leucine rich repeat kinase family, which is the target protein to be ubiquitinated and degraded. In other cases, the term "target protein" is used to refer to a mutant form of LRRK2 protein, such as LRRK protein having one or more mutations selected from the group consisting of G2019S, I2020T, N1437H, R1441G/C/H and Y1699C.
The term "protein targeting moiety" or PTM is used to describe a small molecule that binds to LRRK2 or a mutant form thereof, and can be used to target proteins for ubiquitination and degradation.
The compositions described herein exemplify the use of some of these PTMs.
In any aspect or embodiment described herein, the PTM is a small molecule that binds LRRK 2. For example, in any aspect or embodiment described herein, the PTM is represented by the chemical structure PTM-IA or PTM-IB:
/>
wherein:
R 1 selected from the group consisting of linear or branched C1-C6 alkyl (e.g., isopropyl or t-butyl), optionally substituted C3-C6 cycloalkyl (e.g., optionally substituted C3-C5 cycloalkyl, methylated C3-C5 cycloalkyl),Wherein the method comprises the steps ofIs the point of attachment to M of the PTM), a linear or branched C1-C6 haloalkyl (e.g., a linear or branched C1-C4 haloalkyl), an optionally substituted C3-C6 halocycloalkyl (e.g., a C3-C5 halocycloalkyl), an optionally substituted alkylnitrile (e.g., a C1-C4 alkylnitrile), an optionally substituted C3-C6 cyclic nitrile (e.g., a C3-C5 cyclic nitrile);
R 2 Selected from hydrogen, halogen (e.g., F, cl or Br), C1-C3 alkyl or C1-C3 fluoroalkyl;
X 1 、X 2 、X 3 、X 4 、X 5 、X 6 and X 7 Each independently is C, CH or N, wherein X when CH 1 、X 2 、X 3 And X 4 Each optionally being R 2 Substituted, and X when CH 5 Optionally substituted with C1-C3 alkyl;
X 8 is CH, S or N;
m is CH 2 NH or O;
is an optionally substituted 3-10 membered cycloalkyl, an optionally substituted 3-10 membered heterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered bicycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered spirocycloalkyl, or an optionally substituted 3-10 membered spiroheterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, wherein the heteroatoms are independently selected from N, O and S (e.g., each optionally substituted with one or more (e.g., 1, 2, 3, or 4) substituents), and
PTMrepresents the point of attachment to the linker (L) or ULM.
In any aspect or embodiment described herein, the PTM is represented by the following chemical structure: PTM-IIA, PTM-IIB, PTM-IIIA, PTM-IIIB, PTM-IVA, PTM-IVB, PTM-VA and PTM-VB.
/>
/>
/>
/>
/>
/>
/>
Wherein:
R 1 is a linear or branched C1-C6 alkyl (e.g., isopropyl or tert-butyl), an optionally substituted C3-C6 cycloalkyl (e.g., an optionally substituted C3-C5 cycloalkyl, methylated C3-C5 cycloalkyl), Wherein->Is the point of attachment to M of the PTM), a linear or branched C1-C6 haloalkyl (e.g., a linear or branched C1-C4 haloalkyl), an optionally substituted C3-C6 halocycloalkyl (e.g., a C3-C5 halocycloalkyl), an optionally substituted alkylnitrile (e.g., a C1-C4 alkylnitrile), an optionally substituted C3-C6 cyclic nitrile (e.g., a C3-C5 cyclic nitrile);
R 2 is absent, hydrogen, halogen (e.g., F, cl or Br), OH, C1-C3 alkyl or C1-C3 fluoroalkyl;
X 4 is CH or N;
m is CH 2 NH or O;
is an optionally substituted 3-10 membered cycloalkyl, an optionally substituted 3-10 membered heterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered bicycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered spirocycloalkyl, or an optionally substituted 3-10 membered spiroheterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, wherein the heteroatoms are independently selected from N, O and S (e.g., each optionally substituted with one or more (e.g., 1, 2, 3, or 4) substituents), and
PTMrepresents the point of attachment to a chemical linker group or ULM.
In any aspect or embodiment described herein, Comprising 1 to 4 substitutions, each independently selected from halogen, OH, NH 2 N (C1-C3 alkyl) 2 Linear or branched C1-C4 alkyl (e.g., methyl or ethyl), linear or branched C1-C4 hydroxyalkyl, linear or branched C1-C4 alkoxy, and linear or branched C1-C4 haloalkyl)。/>
In any aspect or embodiment described herein, the PTM is produced by The atoms of the heterocycloalkyl group or substituents thereof are covalently linked to L or ULM.
In any aspect or embodiment described herein,is a 4-7 (e.g., 4, 5, 6, or 7) membered cycloalkyl or heterocycloalkyl containing 1-4 (e.g., 1, 2, 3, or 4) heteroatoms selected from N, O and S, said groups optionally substituted with one or more (e.g., 1, 2, 3, or 4) substituents, each independently selected from halogen, OH, NH 2 N (C1-C3 alkyl) 2 Linear or branched C1-C4 alkyl, linear or branched C1-C4 hydroxyalkyl, linear or branched C1-C4 alkoxy, and linear or branched C1-C4 haloalkyl.
In any aspect or embodiment described herein,is a 4-7 (e.g., 5 or 6) membered cycloalkyl or heterocycloalkyl containing 1-4 (e.g., 1, 2, 3 or 4) heteroatoms selected from N, O and S, said ring being optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents, each independently selected from linear or branched C1-C3 alkyl (e.g., methyl), linear or branched C1-C3 alkoxy (e.g., methoxy) and linear or branched C1-C3 haloalkyl.
In any aspect or embodiment described herein,the method comprises the following steps:
/>
wherein:
R 3 、R 4 and R is 5 Each independently selected from H, halogen, OH, NH 2 N (C1-C3 alkyl) 2 Linear or branched C1-C4 alkyl, linear or branched C1-C4 hydroxyalkyl, linear or branched C1-C4 alkoxy, and linear or branched C1-C4 haloalkyl;
representation->A point of attachment to a 6 membered heteroaryl group of the PTM; and is also provided with
Represents the junction point of PTM with L or ULM, and when not present,/I>Through atoms of cyclic groups (e.g. carbon or nitrogen), R 3 、R 4 Or R is 5 To L or ULM.
In any aspect or embodiment described herein,the method comprises the following steps:
wherein:
R 3 、R 4 and R is 5 Each independently selected from H, halogen, OH, NH 2 N (C1-C3 alkyl) 2 Linear or branched C1-C4 alkyl, linear or branched C1-C4 hydroxyalkyl, linear or branched C1-C4 alkoxy, and linear or branched C1-C4 haloalkyl;
representation->A point of attachment to a 6 membered heteroaryl group of the PTM; and is also provided with
Represents the junction point of PTM with L or ULM, and when not present,/I>Through atoms of cyclic groups (e.g. carbon or nitrogen), R 3 、R 4 Or R is 5 To L or ULM.
In any aspect or embodiment described herein (such as but not limited to aspects or embodiments in the preceding paragraph or in the following paragraphs), The method comprises the following steps: /> In any aspect or embodiment described herein (such as but not limited to aspects or embodiments in the preceding paragraph or in the following paragraphs),the method comprises the following steps: />In any aspect or embodiment described herein (such as but not limited to aspects or embodiments in the preceding paragraph or in the following paragraphs),the method comprises the following steps: /> In any aspect or embodiment described herein (such as but not limited to the aspects or embodiments in the preceding paragraphs), the +_s are provided>The method comprises the following steps: />In any aspect or embodiment described herein (such as but not limited to the aspects or embodiments in the preceding paragraphs), the +_s are provided> The method comprises the following steps:in any aspect or embodiment described herein (such as but not limited to the aspects or embodiments in the preceding paragraphs), the +_s are provided> The method comprises the following steps:in any aspect or embodiment described herein (such as but not limited to the aspects or embodiments in the preceding paragraphs), the +_s are provided>The method comprises the following steps: />
In any aspect or embodiment described herein,the method comprises the following steps: />
Wherein:
R 3 is H or a linear or branched C1-C3 alkyl (e.g., methyl or ethyl);
R 3a is H, halogen, or straight or branched C1-C3 alkyl (e.g., methyl);
R 4 is H or a linear or branched C1-C3 alkyl (e.g., methyl or ethyl);
R 5 is H or a linear or branched C1-C3 alkyl (e.g., methyl);
Representation->A point of attachment to a 6 membered heteroaryl group of the PTM; and is also provided with
Represents the junction point of PTM with L or ULM, and when not present,/I>Through atoms of cyclic groups (e.g. carbon or nitrogen of 6-membered heterocycloalkyl), R 3 、R 4 Or R is 5 To L or ULM.
In any aspect or embodiment described herein,the method comprises the following steps: />
Wherein:
R 3 is H or a linear or branched C1-C3 alkyl (e.g., methyl or ethyl);
R 3a is H, halogen, or straight or branched C1-C3 alkyl (e.g., methyl);
R 4 is H or a linear or branched C1-C3 alkyl (e.g., methyl or ethyl);
R 5 is H or a linear or branched C1-C3 alkyl (e.g., methyl);
representation->A point of attachment to a 6 membered heteroaryl group of the PTM; and is also provided with
Represents the junction point of PTM with L or ULM, and when not present,/I>Through atoms of cyclic groups (e.g. carbon or nitrogen of 6-membered heterocycloalkyl), R 3 、R 4 Or R is 5 To L or ULM.
In any aspect or embodiment described herein,selected from:
/>
/>
wherein R is 3 And R is 4 As defined in any aspect or embodiment described herein.
In any aspect or embodiment described herein,selected from:
/>
wherein R is 3 And R is 4 As defined in any aspect or embodiment described herein.
In any aspect or embodiment described herein,selected from: />
/>
Wherein:
R 3 、R 3a 、R 4 and R is 5 As defined in any aspect or embodiment described herein; and is also provided with
Heterocycloalkyl is substituted by heterocycloalkyl or a substituent thereof (e.g., R 3 、R 3a 、R 4 、R 5 Or methyl) is attached to L or PTM.
In any aspect or embodiment described herein,selected from:
/>
wherein:
R 3 、R 3a 、R 4 and R is 5 As defined in any aspect or embodiment described herein; and is also provided with
Heterocycloalkyl is substituted by heterocycloalkyl or a substituent thereof (e.g., R 3 、R 3a 、R 4 、R 5 Or methyl) is attached to L or PTM.
In any aspect or embodiment described herein, the PTM has the following chemical structure:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
X 2 、X 4 、R 1 、R 2 、R 3 、R 3a 、R 4 and R is 5 As defined in any aspect or embodiment described herein; and is also provided with
PTM is through an atom of heterocycloalkyl A (e.g., carbon or nitrogen of heterocycloalkyl), R 3 、R 4 Or R is 5 To L or ULM.
In any aspect or embodiment described herein, the PTM has the following chemical structure:
/>
/>
/>
/>
/>
/>
wherein: />
X 2 、X 4 、R 1 、R 2 、R 3a 、R 4 And R is 5 As defined in any aspect or embodiment described herein; and is also provided with
PTM (PTM)Representing the point of attachment to L or ULM.
In any aspect or embodiment described herein, R 1 Selected from optionally substituted C3-C5 cycloalkyl and linear or branched C1-C4 alkyl.
In any aspect or embodiment described herein, R 1 Is thatWherein: r is R 1a 、R 1b And R is 1C Each independently is H or a linear or branched C1-C2 alkyl group, each optionally substituted with one or more halogen or nitrile groups; or R is 1a Or R is 1b Together with the carbon to which they are attached, form a C3-C6 cycloalkyl optionally substituted by one or more C1-C3 alkyl groups, nitrile groups or halogens.
In any aspect or embodiment described herein, R 1 Is thatWherein: r is R 1a 、R 1b And R is 1C Each independently is H or a linear or branched C1-C2 alkyl group; or R is 1a Or R is 1b Together with the carbon to which they are attached, form a C3-C6 cycloalkyl group.
In any aspect or embodiment described herein, the PTM is selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
X 2 、X 4 、R 1a 、R 1b 、R 1c 、R 3 、R 3a 、R 4 、R 5 andis defined as described in any aspect or embodiment described herein; and is also provided with
L or ULM is through an atom of the heterocycloalkyl group A (e.g. carbon or nitrogen of the heterocycloalkyl group), R 3 Or R is 4 And (5) connection.
In any aspect or embodiment described herein, the PTM is selected from the group consisting of:
/>
/>
/>
/>
/>
/>
wherein:
X 2 、X 4 、R 1a 、R 1b 、R 1c 、R 3 、R 3a 、R 4 and R is 5 Is defined as described in any aspect or embodiment described herein; and is also provided with
PTM (PTM)Represents the point of attachment to L or ULM, and is in the absence. In any aspect or embodiment described herein, R 1 Selected from-> Wherein->Is the point of attachment to M (e.g., oxygen atom) of PTM.
In any aspect or embodiment described herein, R 2 Is H or F.
In any aspect or embodiment described herein, the PTM has the following chemical structure:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein PTMRepresents the point of attachment to a chemical linker group or ULM.
In any of the aspect embodiments described herein, the PTM has the following chemical structure:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein PTM is covalently linked to L or ULM through an atom of heterocycloalkyl A or a substituent thereof.
In any aspect or embodiment described herein, the PTM has the following chemical structure:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein->Representing the point of attachment to L or ULM.
In any aspect or embodiment described herein, the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
In any aspect or embodiment described herein, the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
In any aspect or embodiment described herein, the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
In any aspect or embodiment described herein, the PTM is represented by:
Wherein said PTMRepresents the point of connection to the L.
In any aspect or embodiment described herein, one or more of the following:
(a)is->/>
Wherein:
R 3 is H or methyl;
R 3a is H, halogen or methyl;
R 4 is H or methyl;
R 5 is H or methyl;
representing a point of connection to the PTM; and is also provided with
Represents the point of attachment to said L and when +.>In the absence of said-> Through N or CH, R of said cyclic group 3 、R 4 Or R is 5 Is attached to the L;
(b)R 2 is H or F; or (b)
(c) A combination thereof.
In any aspect or embodiment described herein, whereinSelected from the group consisting of:
/>
/>
wherein said heterocycloalkyl is attached to said L or said PTM through an atom of said cyclic group or a substituent thereof.
In any aspect or embodiment described herein, the PTM is represented by the following chemical structure:
/>
/>
/>
/>
/>
/>
/>
/>
wherein the method comprises the steps ofRepresents the attachment site of L or CLM.
In any aspect or embodiment described herein, the PTM has the following chemical structure:
/>
in any aspect or embodiment described herein, one or more of the following:
(a)is->
/>
Wherein:
R 3 is H or methyl;
R 3a is H, halogen or methyl;
R 4 is H or methyl;
R 5 is H or methyl;
representing a point of connection to the PTM; and is also provided with
Represents the point of attachment to said L and when +.>In the absence of said-> Through atoms of N or CH, R of said cyclic group 3 、R 4 Or R is 5 Is connected to the L;
(b)R 2 is H or F; or (b)
(c) A combination thereof.
In any aspect or embodiment described herein, whereinSelected from the group consisting of:
/>
/>
wherein said heterocycloalkyl is attached to said L or said PTM through an atom of said cyclic group or a substituent thereof.
In any aspect or embodiment described herein, one or more of the following:
(a)is->
Wherein:
R 3 is H or methyl;
R 3a is H, halogen or methyl;
R 4 is H or methyl;
R 5 is H or methyl;
representing a point of connection to the PTM; and is also provided with
Represents the point of attachment to said L and when +.>In the absence of said-> Through atoms of N or CH, R of said cyclic group 3 Or R is 4 Is connected to the L;
(b)R 2 is H or F; or (b)
(c) A combination thereof.
In any aspect or embodiment described herein,selected from the group consisting of:
wherein the heterocycloalkyl group is attached to L or PTM through an atom of the cyclic group or a substituent thereof.
In any aspect or embodiment described herein, the PTM is represented by the following chemical structure:
/>
/>
wherein->Represents the attachment site of L or CLM.
Therapeutic compositions
The present invention also provides a pharmaceutical composition comprising a therapeutically effective amount of at least one bifunctional compound as described herein in combination with a pharmaceutically acceptable carrier, additive or excipient.
In another aspect, the present description provides therapeutic compositions comprising an effective amount of a compound as described herein, or a salt form thereof, and a pharmaceutically acceptable carrier, additive or excipient, and optionally an additional bioactive agent. Therapeutic compositions achieve targeted protein degradation in a patient or subject (e.g., an animal such as a human) and are useful for treating or ameliorating a disease state or condition modulated by degradation of a target protein. In certain embodiments, therapeutic compositions as described herein may be used to effect protein degradation to treat or ameliorate LRRK2 mediated inflammatory diseases, autoimmune diseases, or cancers. In certain additional embodiments, the disease is idiopathic PD, LRRK2 mutation-associated PD (e.g., PD associated with one or more LRRK2 activating mutations), primary tauopathies (e.g., supranuclear palsy (PSP) or corticobasal degeneration (CBD)), dementia with lewy bodies, crohn's disease, leprosy (e.g., leprosy with type 1 inflammatory response), and/or neuroinflammation.
In an alternative aspect, the present disclosure relates to a method for treating a disease state or ameliorating one or more symptoms of a disease or disorder in a subject in need thereof by degrading LRRK2 protein (e.g., wild-type LRRK2 protein or LRRK2 mutein (e.g., LRRK2 mutein comprising one or more mutations selected from G2019S, I2020T, N1437H, R G/C/H and Y1699C)), the method comprising administering to the patient or subject an effective amount (e.g., a therapeutically effective amount) of at least one compound as described herein, optionally in combination with a pharmaceutically acceptable carrier, additive or excipient, and optionally co-administered with an additional bioactive agent, wherein the composition is effective to treat or ameliorate the disease or disorder or one or more symptoms thereof in the subject. Methods according to the present disclosure may be used to treat certain disease states, conditions, or symptoms, including inflammatory diseases, autoimmune diseases, or cancers, by administering an effective amount of at least one compound described herein. For example, methods according to the present disclosure may be used to treat one or more of the following: parkinson's Disease (PD), idiopathic PD, LRRK2 mutation-associated PD (e.g., PD associated with one or more LRRK2 activating mutations), primary tauopathies (e.g., supranuclear palsy (PSP) or corticobasal degeneration (CBD)), dementia with lewy bodies, crohn's disease, leprosy (e.g., leprosy with type 1 inflammatory response), and neuroinflammation (such as that observed in alzheimer's disease, PD, multiple sclerosis, traumatic brain injury, spinal cord injury, and the like).
The present disclosure also includes pharmaceutical compositions comprising pharmaceutically acceptable salts of the compounds as described herein, particularly acid or base addition salts. The acid used to prepare the pharmaceutically acceptable acid addition salts of the foregoing compounds that can be used in accordance with this aspect is an acid that forms a non-toxic acid addition salt, i.e., a salt containing a pharmacologically acceptable anion, such as hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, acetate, lactate, citrate, acid citrate, tartrate, bitartrate, succinate, maleate, fumarate, gluconate, glucuronate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate [ i.e., 1' -methylene-bis- (2-hydroxy-3-naphthoate) ] and the like.
Pharmaceutically acceptable base addition salts may also be used to produce pharmaceutically acceptable salt forms of the compounds according to the present disclosure. The chemical bases useful as reagents for preparing pharmaceutically acceptable base salts of the compounds of the invention are chemical bases that form non-toxic base salts with such compounds. Such non-toxic base salts include, but are not limited to, base salts derived from such pharmacologically acceptable cations such as alkali metal cations (e.g., potassium and sodium) and alkaline earth metal cations (e.g., calcium, zinc and magnesium), ammonium or water-soluble amine addition salts such as N-methylglucamine- (meglumine), as well as other base salts of lower alkanolammonium and pharmaceutically acceptable organic amines, and the like.
According to the present disclosure, the compounds as described herein may be administered in single or divided doses by oral, parenteral or topical routes. The administration of the active compound may range from continuous (intravenous drip) to several times daily oral administration (e.g., q.i.d.), and may include oral, topical, parenteral, intramuscular, intravenous, subcutaneous, transdermal (which may contain penetration enhancers), buccal, sublingual, intranasal, intraocular, intrathecal, vaginal and suppository administration, and other routes of administration. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Enteric coated oral tablets may also be used to increase the bioavailability of the compound from an oral route of administration. The most effective dosage form will depend on the pharmacokinetics of the particular agent selected, as well as the type, location and severity of the disease, disorder or condition, and the health of the patient. The compounds according to the present disclosure may also be used for intranasal, intratracheal or pulmonary administration as a spray, mist or aerosol administration. Accordingly, the present disclosure also relates to pharmaceutical compositions comprising an effective amount of a compound as described herein, optionally in combination with a pharmaceutically acceptable carrier, additive or excipient. The compounds according to the present disclosure may be administered in immediate release, intermediate release or sustained release or controlled release form. Sustained or controlled release forms are preferably administered orally, but also in suppositories and transdermal or other topical forms. Intramuscular injection of liposomal forms or depot formulations may also be used to control or maintain the release of the compound at the injection site.
The compositions as described herein may be formulated in conventional manner using one or more pharmaceutically acceptable carriers and may also be administered in the form of controlled release formulations. Pharmaceutically acceptable carriers that may be used in these pharmaceutical compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and lanolin, and combinations thereof.
The sterile injectable form of the compositions as described herein may be an aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that can be used are water, ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil (especially in their polyoxyethylated versions). These oil solutions or suspensions may also contain a long chain alcohol diluent or dispersant, such as ph.
The pharmaceutical compositions as described herein may be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of oral tablets, common carriers include lactose and corn starch, as well as other carriers known in the art. For oral administration in capsule form, useful diluents include lactose and corn starch. When an aqueous suspension for oral use is desired, the active ingredient may be combined with emulsifying and suspending agents. If desired, certain sweeteners, flavoring agents or coloring agents may also be added. A lubricant, such as magnesium stearate, is also typically added.
Alternatively, the pharmaceutical compositions as described herein may be used for rectal administration in the form of suppositories. These suppositories may be prepared by mixing the agent with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
The pharmaceutical compositions as described herein may also be administered topically. For topical application, the pharmaceutical compositions may be formulated as transdermal patches, which may be reservoir patches or matrix patches comprising the active compound in combination with one or more carriers, buffers, absorption enhancers and providing continuous administration for 1 day to two weeks.
Alternatively, the pharmaceutical compositions of the present disclosure may be formulated in a suitable paste containing the active ingredient suspended or dissolved in one or more carriers. Carriers for topical application of the compounds of the present disclosure include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compounds, emulsifying wax and water.
Alternatively, the pharmaceutical compositions of the present disclosure may be formulated as a suitable lotion or cream containing the active component suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetostearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
Alternatively, the pharmaceutical compositions of the present disclosure may be formulated for ophthalmic use. For example, the pharmaceutical composition may be formulated as a micronized suspension in isotonic, pH adjusted sterile saline, or, preferably, as a solution in isotonic, pH adjusted sterile saline, with or without the use of a preservative such as benzalkonium chloride. Alternatively, for ophthalmic applications, the pharmaceutical compositions may be formulated as ointments such as petrolatum.
The pharmaceutical compositions as described herein may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well known in the art of pharmaceutical formulation, and may be prepared as solutions in physiological saline using benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
The amount of active pharmaceutical ingredient in a pharmaceutical composition as described herein that can be combined with a carrier material to produce a single dosage form will vary depending upon the condition of the subject and the disease, disorder or condition being treated, the particular mode of administration, and the condition of the subject. Preferably, the composition should be formulated to contain between about 0.05 mg and about 750 mg or more, more preferably about 1 mg to about 600 mg, and even more preferably about 10 mg to about 500 mg of the active ingredient, alone or in combination with another compound according to the present disclosure.
It will also be appreciated that the specific dosage and treatment regimen for any particular patient will depend upon a number of factors including the activity and bioavailability of the particular compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, the judgment of the treating physician, and the severity of the particular disease or condition undergoing therapy.
A patient or subject in need of treatment with a compound according to the methods described herein may be treated by administering to the patient (subject) an effective amount of the compound according to the present disclosure, optionally alone or in combination with another known therapeutic agent in a pharmaceutically acceptable carrier or diluent, depending on the pharmaceutically acceptable salt or solvate of the compound according to the present disclosure.
In certain aspects, the active compound is combined with a pharmaceutically acceptable carrier or diluent in an amount sufficient to deliver to the patient a therapeutically effective amount for the desired indication without undue serious toxic effects on the patient being treated. Preferred dosages of the active compound for all of the disorders mentioned herein range from about 10 nanograms per kilogram (ng/kg) to 300 milligrams per kilogram (mg/kg), preferably from 0.1 to 100mg/kg per day, more typically from 0.5 to about 25 mg/kg of recipient/patient body weight per day. Typical topical dosages will range from 0.01 to 5% wt/wt in a suitable carrier.
In certain aspects, the compounds are conveniently administered in any suitable unit dosage form, including, but not limited to, dosage forms containing less than 1 milligram (mg), 1mg to 3000mg, or 5mg to 500mg of active ingredient per unit dosage form. An oral dosage of about 25mg to 250mg is generally convenient.
In certain aspects, the active ingredient is preferably administered to achieve a peak plasma concentration of the active compound of about 0.00001 to 30 millimoles (mM), preferably about 0.1 to 30 micromoles (μM). This can be achieved, for example, by intravenous injection of a solution or formulation of the active ingredient, optionally administered in saline or an aqueous medium or as a bolus (bolus) of the active ingredient. Oral administration may also be suitable to produce an effective plasma concentration of the active agent.
The concentration of the active compound in the pharmaceutical composition will depend on the absorption, distribution, inactivation, and excretion rates of the drug as well as other factors known to those of skill in the art. It should be noted that the dosage value will also vary with the severity of the condition to be alleviated. It will be further understood that the specific dosage regimen for any particular subject will be adjusted over time according to the individual needs and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions. The active ingredient may be administered at one time or may be divided into a plurality of smaller doses for administration at different time intervals.
The oral composition will typically include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound or prodrug derivative thereof may be combined with excipients and used in the form of tablets, troches or capsules. Pharmaceutically compatible binders and/or adjuvant materials may be included as part of the composition.
Tablets, pills, capsules, troches and the like may contain any of the following ingredients or compounds of similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; excipients, such as starch or lactose; dispersants such as alginic acid, primogel or corn starch; lubricants, such as magnesium stearate or Sterotes; glidants such as colloidal silicon dioxide; sweeteners such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a fatty oil. In addition, the dosage unit form may contain various other materials that alter the physical form of the dosage unit, such as a coating of sugar, shellac, or enteric agents.
The active compound or pharmaceutically acceptable salt thereof can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum, or the like. Syrups may contain, in addition to the active compounds, sucrose (as sweetener) and certain preservatives, dyes and colorants and flavors.
The active compound or pharmaceutically acceptable salt thereof may also be admixed with other active materials which do not impair the desired effect, or with materials which supplement the desired effect, such as anti-cancer agents described herein, and the like. In certain preferred aspects of the present disclosure, one or more compounds according to the present disclosure are co-administered with another bioactive agent as further described herein, such as an anticancer agent or a wound healing agent (including antibiotics).
Solutions or suspensions for parenteral, intradermal, subcutaneous, or topical application may include the following components: sterile diluents such as water for injection, saline solutions, fixed oils, polyethylene glycols, glycerol, propylene glycol or other synthetic solvents; antimicrobial agents such as benzyl alcohol or methylparaben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediamine tetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for modulating tonicity such as sodium chloride or dextrose. Parenteral formulations may be packaged in ampules, disposable syringes or multiple dose vials made of glass or plastic.
If administered intravenously, the preferred carrier is physiological saline or Phosphate Buffered Saline (PBS).
In any aspect or embodiment, the active compound is prepared with a carrier that will prevent rapid elimination of the compound from the body, such as a controlled release formulation comprising an implant and a microencapsulated delivery system. Biodegradable biocompatible polymers such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters and polylactic acid may be used. Methods for preparing such formulations will be apparent to those skilled in the art.
The liposome suspension may also be a pharmaceutically acceptable carrier. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811, which is incorporated herein by reference in its entirety. For example, a liposome formulation can be prepared by: the appropriate lipid or lipids (such as stearoyl phosphatidylethanolamine, stearoyl phosphatidylcholine, arachidoyl phosphatidylcholine, and cholesterol) are dissolved in an inorganic solvent and then evaporated, leaving a dry lipid film on the surface of the container. An aqueous solution of the active compound is then introduced into the container. The vessel is then vortexed by hand to detach the lipid material from the sides of the vessel and disperse the lipid aggregates, thereby forming a liposome suspension.
Therapeutic method
In another aspect, the present specification provides a method of treatment comprising administering an effective amount of a compound as described herein, or a salt form thereof, and a pharmaceutically acceptable carrier. The methods of treatment are useful for effecting protein degradation in a patient or subject (e.g., an animal such as a human) in need thereof for treating or ameliorating a disease state, disorder, or related symptom treatable by targeted protein degradation.
The terms "treatment", "treatment" and the like as used herein refer to any action that may provide a benefit to a patient to whom a compound of the invention may be administered, including treatment of any disease state, disorder or symptom associated with a protein to which the compound of the invention binds. The foregoing illustrates disease states or conditions, including cancer, that may be treated using compounds according to the present disclosure.
The present specification provides methods of treatment for effecting degradation of a protein of interest for treating or ameliorating a disease, such as Parkinson's Disease (PD), primary tauopathies, lewy body dementia, crohn's disease, leprosy disease, and/or neuroinflammation (such as described in any aspect or embodiment, the disease is idiopathic PD, LRRK2 mutation-related PD (e.g., PD associated with one or more LRRK2 activating mutations), PSP, CBD, leprosy with type 1 inflammatory response, alzheimer's disease, PD, multiple sclerosis, traumatic brain injury, and/or spinal cord injury).
In further embodiments, the present specification provides methods for treating or ameliorating a disease, disorder, or symptom thereof in a subject or patient (e.g., an animal such as a human), the methods comprising administering to a subject in need thereof a composition comprising an effective amount (e.g., a therapeutically effective amount) of a compound as described herein or a salt form thereof and a pharmaceutically acceptable excipient, carrier, adjuvant, another bioactive agent, or combination thereof, wherein the composition is effective to treat or ameliorate the disease or disorder, or symptom thereof in the subject.
In another aspect, the present specification provides methods for identifying the effect of degrading a protein of interest in a biological system using a compound according to the present disclosure.
In another aspect, the present specification provides a method for preparing a molecule that can cause degradation of intracellular LRRK2, the method comprising the steps of: (i) providing a small molecule that binds to LRRK2 or a mutant form thereof; (ii) Providing an E3 ubiquitin ligase binding moiety (ULM), preferably CLM, such as thalidomide, pomalidomide, lenalidomide, or an analog thereof; and (iii) covalently coupling the small molecule of step (i) to the ULM of step (ii) via a chemical linking group (L) to form a compound that binds to the cereblon E3 ubiquitin ligase and the LRRK2 protein and/or mutated form in the cell such that the cereblon E3 ubiquitin ligase approaches and ubiquitinates the LRRK2 protein bound thereto, whereby the ubiquitinated LRRK2 is subsequently degraded.
In another aspect, the present specification provides a method for detecting whether a molecule can trigger degradation of LRRK2 protein in a cell, the method comprising the steps of: (i) Providing a molecule to be detected that has the ability to trigger a decrease in LRRK2 protein in a cell, said molecule comprising the structure: CLM-L-PTM, wherein CLM is a cereblon E3 ubiquitin ligase binding moiety capable of binding a cereblon E3 ubiquitin ligase in the cell, said CLM is thalidomide, pomalidomide, lenalidomide, or an analog thereof; PTM is a protein targeting moiety which is a small molecule that binds to LRRK2 and/or mutated LRRK forms thereof, said LRRK2 having at least one lysine residue that can be ubiquitinated by the cereblon E3 ubiquitin ligase of CLM that binds to this molecule; and L is a chemical linking group that covalently links CLM with PTM to form a molecule; (ii) Incubating cells expressing LRRK2 protein in the presence of the molecule of step (i); and (iii) detecting whether the LRRK2 protein in the cell is degraded.
In any aspect or embodiment described herein, the small molecule capable of binding LRRK2 is a small molecule that binds LRRK 2. In certain embodiments, small molecules that bind LRRK2 are as described herein.
In another aspect of the treatment, the present disclosure provides a method of treating a human patient in need of such treatment for a disease state, disorder or condition that is causally related to LRRK2 and/or mutant forms, expression, overexpression, mutation, aggregation, accumulation, misfolding or deregulation of LRRK2, wherein degradation of LRRK2 protein will produce a therapeutic effect in the patient, the method comprising administering to the patient an effective amount of a compound according to the present disclosure, optionally in combination with another bioactive agent.
In another aspect of the treatment, the present disclosure provides a method of treating a human patient in need of such treatment for a disease state, disorder or condition that is causally related to α -synuclein expression, overexpression, mutation, aggregation, accumulation, misfolding or deregulation, wherein degradation of LRRK2 protein and/or mutant forms thereof will produce a therapeutic effect in the patient, the method comprising administering to the patient an effective amount of a compound according to the present disclosure, optionally in combination with another bioactive agent.
In another aspect of the treatment, the present disclosure provides a method of treating a human patient in need of such treatment for a disease state, disorder or symptom having a causal relationship with α -synuclein expression, overexpression, mutation, aggregation, misfolding or deregulation, wherein degradation of LRRK2 protein and/or mutant forms thereof will produce a therapeutic effect in the patient, the method comprising administering to the patient an effective amount of a compound according to the present disclosure, optionally in combination with another bioactive agent.
In another aspect of the treatment, the present disclosure provides a method of treating a human patient in need of such treatment for a disease state, disorder or symptom having a causal relationship with Tau expression, overexpression, mutation, aggregation, misfolding or deregulation, wherein degradation of LRRK2 protein and/or mutant forms thereof will produce a therapeutic effect in the patient, the method comprising administering to the patient an effective amount of a compound according to the present disclosure, optionally in combination with another bioactive agent.
The disease state, disorder or condition may be caused by microbial factors or other exogenous factors such as viruses, bacteria, fungi, protozoa or other microorganisms, or may be a disease state caused by the expression, overexpression, mutation, misfolding or deregulation of proteins that lead to the disease state, disorder or condition.
In another aspect, the present disclosure provides a method of treating or ameliorating at least one symptom of a disease or disorder in a subject, the method comprising the steps of:
providing a subject identified as having symptoms of a disease or disorder causally related to expression, overexpression, mutation, misfolding or deregulation of LRRK2 protein and/or a mutant form thereof in the subject, and treating or ameliorating the symptoms of the disease or disorder by degrading LRRK2 protein and/or a mutant form thereof in cells of the subject; and administering to the subject a therapeutically effective amount of a compound comprising a small molecule of the invention such that the LRRK2 protein and/or mutant forms thereof are degraded, thereby treating or ameliorating at least one symptom of the disease or disorder in the subject.
The term "disease state or condition" is used to describe any disease state or condition in which overexpression, mutation, misfolding or deregulation of protein expression (e.g., an increase in the amount of protein expressed in a patient) occurs, and in which LRRK2 protein and/or mutant forms thereof degrade to reduce or stabilize the level of LRRK2 protein (whether mutated or not) in a patient, providing beneficial treatment or symptomatic relief to a patient in need thereof. In some cases, the disease state, disorder or condition may be cured.
The disease states, disorders, or symptoms that may be treated using compounds according to the present disclosure include, for example, parkinson's Disease (PD), idiopathic PD, LRRK2 mutation-related PD (e.g., PD associated with one or more LRRK2 activating mutations), primary tauopathies (e.g., supranuclear palsy (PSP) or corticobasal degeneration (CBD)), lewy body dementia, crohn's disease, leprosy (e.g., leprosy with type 1 inflammatory response), and/or neuroinflammation (such as that observed in alzheimer's disease, PD, multiple sclerosis, traumatic brain injury, spinal cord injury, and the like).
The term "bioactive agent" is used to describe agents other than the compounds according to the present disclosure that are used in combination with the compounds of the present invention as bioactive agents to help achieve the desired treatment, inhibition, and/or prevention/control using the compounds of the present invention. Preferred bioactive agents for use herein include those having similar pharmacological activity to that of the compounds of the invention used or administered, and include, for example, anticancer agents, antiviral agents, including, inter alia, anti-HIV and anti-HCV agents, antimicrobial agents, antifungal agents, and the like.
The term "additional anti-autoimmune disease agent" is used to describe a therapeutic agent against an autoimmune disease that may be combined with a compound according to the present disclosure to treat an autoimmune disease. Such agents include, for example, infliximab, tofacitinib, baratinib, secukinumab, adalimumab, etanercept, golimumab, cetuzumab (certolizumab pepol), antiproliferative agents (e.g., mycophenolate mofetil (mycophenolate mofetil)) and corticosteroids.
The term "pharmaceutically acceptable derivative" is used throughout the specification to describe any pharmaceutically acceptable prodrug form (such as an ester, amide or other prodrug group) that provides, directly or indirectly, a compound of the invention or an active metabolite of the compound of the invention after administration to a patient.
Examples
Abbreviations (abbreviations)
ACN acetonitrile
AcOH acetic acid
Boc
dba dibenzylidene acetone
DBU 1, 8-diazabicyclo [5.4.0] undec-7-ene
DCM dichloromethane
DMA dimethylacetamide
DME dimethoxyethane
DMF dimethylformamide
DMSO dimethyl sulfoxide
DMAC/DMA dimethylacetamide
DIEA N, N-diisopropylethylamine
EDTA ethylenediamine tetraacetic acid
EtOAc/EA ethyl acetate
EtOH ethanol
FA formic acid
HPLC high pressure liquid chromatography
Hz hertz
IBX 2-iodate benzoic acid
Lithium aluminium hydride LAH
LCMS liquid chromatography/mass spectrometry
LiHMDS lithium bis (trimethylsilyl) amide
MHz megahertz (MHz)
NBS N-bromosuccinimide
NCS N-chlorosuccinimide
NMR nuclear magnetic resonance
NMP N-methyl-2-pyrrolidone
MeOH methanol
MPLC medium pressure liquid chromatography
MTBE methyl tert-butyl ether
PE Petroleum ether
Psi pound force per square inch
RT or r.t. room temperature
SFC supercritical fluid chromatography
TEA triethylamine
THF tetrahydrofuran
TFA trifluoroacetic acid
TLC thin layer chromatography
TMS trimethylsilyl group
* Number of repetitions
General synthetic method
The synthetic implementation and optimization of bifunctional molecules as described herein may be performed in a stepwise or modular manner. For example, if no suitable ligand is immediately available, identifying compounds that bind to the target protein (i.e., LRRK 2) may involve high-throughput or medium-throughput screening activities. It is not uncommon that the initial ligand requires iterative design and optimization loops to improve sub-optimal aspects as identified by data from appropriate in vitro and pharmacological and/or ADMET assays. Part of the optimization/SAR activity will be the tolerant substitution of the probe ligand and possibly the position of the appropriate site for attachment of the chemical linking group referred to herein before. Where crystallographic or NMR structural data is available, these data can be used to focus this synthetic effort.
In a very similar manner, ligands for E3 ligase can be identified and optimized.
With PTM and ULM (e.g., CLM), one skilled in the art can combine them with or without one or more chemical linking groups using known synthetic methods. One or more chemical linking groups of a range of compositions, lengths and flexibilities can be synthesized and functionalized so that the PTM and ULM groups can be sequentially attached to the distal end of the linker. Thus, libraries of bifunctional molecules can be realized and analyzed in vitro and in vivo pharmacological and ADMET/PK studies. As with the PTM and ULM groups, the final bifunctional molecule may be subjected to iterative design and optimization cycles to identify molecules with desired properties.
In some cases, protecting group strategies and/or Functional Group Interconversions (FGIs) may be required to facilitate the preparation of the desired materials. Such chemistry is well known to synthetic organic chemists, and many of them can be found in texts such as "Greene's Protective Groups in Organic Synthesis" Peter g.m. wuts and the thodora w.greene (Wiley) and "Organic Synthesis: the Disconnection Approach" Stuart Warren and Paul Wyatt (Wiley).
Synthesis procedure
General synthetic scheme
Scheme 1:
scheme 2:
scheme 3:
x represents a suitable leaving group (e.g., OTs, OMs, cl, br, etc.)
Y represents primary or secondary amine or primary or secondary alcohol
M represents a metallized version of TLM (Na + 、cs + 、Li + Etc
PG represents a suitable protecting group
Exemplary Synthesis of the intermediates 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane and 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methylpiperazin-1-yl ] pyrimidin-4-yl ] -1H-indazole
Step 1
At 0 ℃ at N 2 To a solution of 2-bromo-4-fluoro-1-nitro-benzene (16.78 g,76.28mmol,1.1 eq.) and 1-methylcyclopropanol (5 g,69.34mmol,1 eq.) in DMF (160 mL) was added NaH (4.16 g,104.01mmol, 60% in mineral oil, 1.5 eq.) in one portion. The mixture was then heated to 20 ℃ and stirred for 4 hours. TLC showed new spots. The residue was poured into water (200 mL) and stirred for 10min. The aqueous phase was extracted with ethyl acetate (3X 300 mL). The combined organic phases were washed with brine (2X 200 mL) and dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (100-200 mesh silica gel, 0% -2% ethyl acetate/petroleum ether) to give 2-bromo-4- (1-methylcyclopropoxy) -1-nitro-benzene (14.3 g,52.56mmol,75.79% yield) as a yellow oil.
Step 2
To 2-bromo-4- (1-methylcyclopropoxy) -1-nitro-benzene (14.3 g,52.56mmol,1 eq.) K at 20deg.C 2 CO 3 (14.53 g,105.11mmol,2 eq.) and Cs 2 CO 3 (17.12 g,52.56mmol,1 eq.) to a mixture of 1, 4-dioxane (100 mL) was added 2,4, 6-trimethyl-1,3,5,2,4,6-trioxadiborane (32.99 g,131.39mmol,36.73mL, 50% purity in EtOAc, 2.5 eq.) and Pd (PPh 3 ) 4 (6.07 g,5.26mmol,0.1 eq.) followed by heating to 100℃and stirring for 16 hours gave a yellow solution. TLC showed the reaction was complete. The reaction was cooled to 20 ℃ and concentrated under vacuum. PE: E is added to the residueto (10:1, 100 mL), and the mixture was filtered through a pad of silica. The pad was washed with petroleum ether: etOAc (10:1, 1000 mL) solvent. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -1% ethyl acetate/petroleum ether) to give 2-methyl-4- (1-methylcyclopropoxy) -1-nitro-benzene (11 g, crude) as a yellow oil.
Step 3
At 20 ℃ at N 2 To a mixture of 2-methyl-4- (1-methylcyclopropoxy) -1-nitro-benzene (11 g,53.08mmol,1 eq.) in EtOH (100 mL) was added 10% Pd/C (4 g,5.31mmol,0.1 eq.) and ammonium formate (40.17 g,636.99mmol,12 eq.) in one portion. The mixture was stirred at 20 ℃ for 2h to give a black mixture. TLC showed the reaction was complete. The mixture was filtered through a pad of silica gel, washed with EtOAc (3×200 mL) and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -10% ethyl acetate/petroleum ether) to give 2-methyl-4- (1-methylcyclopropoxy) aniline (9.8 g, crude) as a red oil.
Step 4
At 0 ℃ at N 2 Downward 2-methyl-4- (1-methylcyclopropoxy) aniline (9.8 g,55.29mmol,1 eq.) and Et 3 N (13.99 g,138.23mmol,19.24mL,2.5 eq.) Ac was added in one portion to a mixture of DCM (100 mL) 2 O (11.29 g,110.58mmol,10.36mL,2 eq.). The mixture was stirred at 0 ℃ for 30min, then heated to 20 ℃ and stirred for 16 hours. TLC showed the reaction was complete. The reaction was taken up with saturated NaHCO 3 The aqueous solution (30 mL) was quenched to adjust ph=7-8 and extracted with DCM (3×50 mL). The combined organic phases were washed with brine (3X 50 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (20% -40%Ethyl acetate/petroleum ether) to give N- [ 2-methyl-4- (1-methylcyclopropoxy) phenyl ] as a yellow oil]Acetamide (9.3 g,42.41mmol,76.71% yield).
Step 5
To N- [ 2-methyl-4- (1-methylcyclopropoxy) phenyl at 20 DEG C]To a solution of acetamide (9.3 g,42.41mmol,1 eq.) in toluene (100 mL) was added KOAc (6.24 g,63.62mmol,1.5 eq.) and Ac 2 O (19.92 g,195.09mmol,18.27mL,4.6 eq.) the solution was heated to 80℃followed by dropwise addition of 3-methylbutyl nitrite (19.87 g,169.65mmol,22.84mL,4 eq.). After the addition, the mixture was stirred at 80 ℃ for 2h. TLC showed the reaction was complete. The reaction was then filtered, the wet cake was washed with EtOAc (70 mL), and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% ethyl acetate/petroleum ether) to give 1- [5- (1-methylcyclopropoxy) indazol-1-yl as a yellow solid ]Ethanone (8 g, crude).
Step 6
To 1- [5- (1-methylcyclopropoxy) indazol-1-yl at 20 DEG C]To a mixture of ethanone (8 g,34.74mmol,1 eq.) in MeOH (80 mL) was added NH in one portion 3 (g /) MeOH (7M, 24.82mL,5 eq.). The mixture was stirred at 20 ℃ for 2 hours to give a yellow solution. TLC showed the reaction was complete. The solution was concentrated in vacuo to give 5- (1-methylcyclopropoxy) -1H-indazole (7.8 g, crude) as a yellow solid.
Step 7
To 5- (1-methyl) at 20 DEG CTo a mixture of the propyloxy-1H-indazole (7.8 g,41.44mmol,1 eq.) in THF (80 mL) were added N-dicyclohexylmethylamine (10.52 g,53.87mmol,1.3 eq.) and SEM-Cl (8.29 g,49.73mmol,8.80mL, 1.2 eq.) in one portion. The mixture was stirred at 20 ℃ for 16 hours to give an orange solution. TLC showed the reaction was complete. The residue was poured into water (60 mL). The aqueous phase was extracted with ethyl acetate (3X 50 mL). The combined organic phases were washed with brine (2X 50 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% ethyl acetate/petroleum ether) to give trimethyl- [2- [ [5- (1-methylcyclopropoxy) indazol-2-yl ] as a yellow oil ]Methoxy group]Ethyl group]Silane (5.4 g,16.96mmol,40.92% yield).
Step 8
At-70 ℃ at N 2 Downward trimethyl- [2- [ [5- (1-methylcyclopropoxy) indazol-2-yl ]]Methoxy group]Ethyl group]To a mixture of silane (4.36 g,13.70mmol,5.32e-1 eq.) in THF (6 mL) was added n-BuLi (2.5M, 13.40mL,1.3 eq.) dropwise. The mixture was then stirred at-20℃for 1h and ZnCl was added dropwise at-70 ℃ 2 Solution (0.7M, 55.20mL,1.5 eq.). The mixture was stirred at-40℃for 1h. 4, 6-dichloropyrimidine (4.22 g,28.34mmol,1.1 eq.) and Pd (PPh) 3 ) 4 A mixture of (1.49 g,1.29mmol,0.05 eq.) in THF (4 mL) was stirred at 20deg.C for 1h and added to the solution. The cold bath was removed and the mixture was stirred at 20 ℃ for 16h to give a yellow solution. TLC showed some starting material remained while some new spots formed. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (3X 20 mL). The combined organic phases were washed with brine (2X 20 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% ethyl acetate/petroleum ether) to give 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1) as a yellow oil -methylcyclopropoxy) indazol-2-yl]Methoxy group]Ethyl-trimethyl-silane (2.9 g, crude).
Step 9
To 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl]Methoxy group]Et is added in one portion to a mixture of ethyl-trimethyl-silane (500 mg,1.16mmol,1 eq.) and tert-butyl (2S) -2-methylpiperazine-1-carboxylate (697.02 mg,3.48mmol,3 eq.) in DMSO (5 mL) 3 N (704.34 mg,6.96mmol,968.82uL,6 eq.) the mixture was then stirred at 100deg.C for 1h. TLC showed the reaction was complete. The mixture was cooled to 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (3X 5 mL). The combined organic phases were washed with brine (2X 5 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl as a yellow oil]Pyrimidin-4-yl]Piperazine-1-carboxylic acid tert-butyl ester (802 mg, crude).
Step 10
To (2S) -2-methyl-4- [6- [5- [ (1-methylcyclopropyl) methyl ] at 25 DEG C]-2- (2-trimethylsilylethoxymethyl) indazol-3-yl]Pyrimidin-4-yl]To a mixture of tert-butyl piperazine-1-carboxylate (803 mg,1.35mmol,1 eq.) in DCM (5 mL) was added TFA (771.25 mg,6.76mmol,500.81uL,5 eq.) in one portion. The mixture was stirred at 25℃for 16 hours. HCl (4M, 338.20uL,1 eq.) in MeOH (5 mL) was added at 25deg.C followed by heating to 60deg.C and stirring for 0.5 h. LCMS showed the reaction was complete. The mixture was cooled to 20 ℃. Pouring the residue into NaHCO 3 (5 mL) to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (3X 10 mL). The combined organic phases were washed with brine (2X 10 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -40% ethyl acetate/MeOH) to give 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methylpiperazin-1-yl as a yellow solid]Pyrimidin-4-yl]-1H-indazole (450 mg,1.18mmol,87.41% yield, 95.77% purity).
Exemplary Synthesis of intermediate 5- (1-methylcyclopropoxy) -3- (6-piperazin-1-ylpyrimidin-4-yl) -1H-indazole
Step 1
To 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl]Methoxy group]To a solution of ethyl-trimethyl-silane (500 mg,1.16mmol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester (648.20 mg,3.48mmol,3 eq.) in DMSO (5 mL) was added Et in one portion 3 N (704.34 mg,6.96mmol,968.82uL,6 eq.) the mixture was then stirred at 100deg.C for 1h. TLC showed the reaction was complete. The mixture was cooled to 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (3X 5 mL). The combined organic phases were washed with brine (2X 5 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl as a yellow oil ]Pyrimidin-4-yl]Piperazine-1-carboxylic acid tert-butyl ester (812 mg, crude).
Step 2
At 25℃to 4- [6- [5- [ (1-methylcyclopropyl) methyl)]-2- (2-trimethylsilylethoxymethyl) indazol-3-yl]Pyrimidin-4-yl]To a mixture of tert-butyl piperazine-1-carboxylate (812 mg,1.40mmol,1 eq.) in DCM (5 mL) was added TFA (799.77 mg,7.01mmol,519.33uL,5 eq.) in one portion and stirred at 25℃for 16h. HCl (g) (4 m,5.26mL,15 equivalents), followed by heating to 60 ℃ and stirring for 0.5 hours. The mixture was cooled to 20 ℃. LCMS showed the reaction was complete. Pouring the residue into NaHCO 3 (5 mL) to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (3X 10 mL). The combined organic phases were washed with brine (2X 10 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -40% ethyl acetate/MeOH) to give 5- (1-methylcyclopropoxy) -3- (6-piperazin-1-ylpyrimidin-4-yl) -1H-indazole as a white solid (290 mg,827.59umol,58.99% yield).
Exemplary Synthesis of intermediate 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane
Step 1
To a solution of 5- (1-methylcyclopropoxy) -1H-indazole (1 g,4.45mmol,1 eq, HCl) in DMF (10 mL) was added I2 (2.26 g,8.90mmol,1.79mL,2 eq) and KOH (749.18 mg,13.35mmol,3 eq). After the addition, the reaction mixture was stirred at 25 ℃ for 3h. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=5:1) showed a major new spot. The reaction mixture was quenched with saturated Na2S2O3 (20 mL) and extracted with ethyl acetate (2×20 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 15% ethyl acetate/petroleum ether) to give 3-iodo-5- (1-methylcyclopropoxy) -1H-indazole (1.4 g,4.35mmol,97.84% yield, 97.7% purity) as a pale yellow oil.
Step 2
To a mixture of 3-iodo-5- (1-methylcyclopropoxy) -1H-indazole (1.2 g,3.82mmol,1 eq.) in THF (15 mL) was added N-cyclohexyl-N-methyl-cyclohexylamine (2.24 g,11.46mmol,2.43mL,3 eq.) and SEM-Cl (1.27 g,7.64mmol,1.35mL,2 eq.). After the addition, the reaction mixture was stirred at 25 ℃ for 2 hours to give a white suspension. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=5:1) showed several new spots. The reaction mixture was diluted with water (150 mL) and extracted with ethyl acetate (150 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 12% ethyl acetate/petroleum ether) to give 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (630 mg,1.36mmol,35.55% yield, 95.8% purity) as a pale yellow oil.
Exemplary Synthesis of intermediate 2- (2, 6-dioxopiperidin-3-yl) -5- (piperazin-1-yl) isoindoline-1, 3-dione hydrochloride
Step 1
To 5-fluoroisobenzofuran-1, 3-dione (1 g,6.02mmol,1 eq.) and 3-aminopiperidine-2, 6-dione HCl salt (1.49 g,9.03mmol,1.5 eq.) in CH 3 KOAc (1.18 g,12.04mmol,2 eq.) was added to a solution of COOH (10 mL). After addition, the reaction mixture was stirred at 120 ℃ for 12 hours. The mixture was diluted with water (40 mL). The mixture was filtered and the filter cake was washed with water (100 mL) to give 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (1.4 g,5.07mmol,84.19% yield) as a black solid.
Step 2
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (1.15 g,4.16mmol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester (852.97 mg,4.58mmol,1.1 eq.) in NMP (10 mL) was added DIEA (1.61 g,12.49mmol,2.18mL,3 eq.). The sealed tube was heated under microwaves at 140 ℃ for 2 hours. The mixture was combined with the second batch, diluted with water (50 mL) and extracted with ethyl acetate (2×50 mL). The combined organic phases were washed with saturated brine (2×30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 50% ethyl acetate/petroleum ether) to give 4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazine-1-carboxylic acid tert-butyl ester (1.4 g,3.16 mmol) as a yellow solid. The average yield of the combined batches was 62.49%.
Step 3
To a solution of 4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazine-1-carboxylic acid tert-butyl ester (1.2 g,2.71mmol,1 eq.) in MeOH (10 mL) was added HCl/dioxane (4 m,2.00mL,2.95 eq.). After the addition, the reaction solution was stirred at 65 ℃ for 1h. The reaction solution was combined with the second batch. The mixture was concentrated under reduced pressure to give 2- (2, 6-dioxopiperidin-3-yl) -5- (piperazin-1-yl) isoindoline-1, 3-dione hydrochloride (1.1 g, crude) as a yellow solid. The average yield of the combined batches was 91.04%.
Exemplary Synthesis of intermediate 4-methylbenzenesulfonic acid [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ester
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (100 mg,362.03umol,1 eq.) and 4-piperidinylmethanol (83.39 mg,724.06umol,2 eq.) in DMSO (2 mL) was added DIEA (140.37 mg,1.09mmol,189.18uL,3 eq.) at 20deg.C in one portion. The mixture was stirred at 100℃for 3h. TLC (DCM: meoh=10:1, rf=0.36) showed the reaction was complete. The mixture was cooled to 20 ℃. Pouring the residue into NaHCO 3 (10mL)To adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (3X 10 mL). The combined organic phases were washed with brine (2X 10 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% MeOH/DCM) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (hydroxymethyl) -1-piperidinyl as a yellow gum]Isoindoline-1, 3-dione (120 mg,283.18umol,78.22% yield, 87.640% purity).
Step 2
At 0 ℃ at N 2 Downward 2- (2, 6-dioxo-3-piperidyl) -5- [4- (hydroxymethyl) -1-piperidyl]To a solution of isoindoline-1, 3-dione (120 mg,323.11umol,1 eq.) and TEA (98.09 mg,969.34umol,134.92uL,3 eq.) in DCM (5 mL) was added TsCl (27.35 mg,387.74umol,1.2 eq.) in one portion. The mixture was stirred at 20 ℃ for 20 hours to give a yellow solution. TLC (DCM: meoh=10:1, rf=0.45) showed the reaction was complete. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (3X 5 mL). The combined organic phases were washed with brine (2X 5 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -50% ethyl acetate/petroleum ether (5 min), 50-100 ethyl acetate/petroleum ether (10 min)) to give 4-methylbenzenesulfonic acid [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] as a yellow solid ]-4-piperidinyl]Methyl ester (160 mg,228.91umol,70.85% yield, 75.194% purity).
Exemplary Synthesis of intermediate 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde
Step 1
At 25 deg.C to 2 # -To a solution of 2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (300 mg,1.09mmol,1 eq.) and 4- (dimethoxymethyl) piperidine (207.52 mg,1.30mmol,1.2 eq.) in DMSO (5 mL) was added DIEA (421.11 mg,3.26mmol,567.53uL,3 eq.) followed by stirring the reaction at 100deg.C for 1h to give a brown solution. TLC (dichloromethane: methanol=10:1, rf=0.5) showed the desired product was detected. Pouring the reaction mixture into H 2 O (5 mL). The mixture was extracted with ethyl acetate (10 ml x 3). The organic phase was washed with brine (10 mL), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by silica gel chromatography (column height: 250mm, diameter: 100mm,100-200 mesh silica gel, petroleum ether/ethyl acetate=100/1, 1/1) to give 5- [4- (dimethoxymethyl) -1-piperidinyl as a yellow solid]-2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (326 mg,580.69umol,53.47% yield, 74% purity).
Step 2
To 5- [4- (dimethoxymethyl) -1-piperidinyl]To a solution of 2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (140 mg,336.99umol,1 eq.) in THF (3 mL) was added HCl (2M, 3mL,17.80 eq.). After the addition, the reaction solution was stirred at 70 ℃ for 1h. LCMS showed the desired MS. The reaction solution was passed through saturated NaHCO 3 (pH about 8) and extracted with ethyl acetate (3X 15 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure to give 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl as a yellow solid]Piperidine-4-carbaldehyde (124 mg,302.14umol,89.66% yield, 90% purity). The crude product was used directly.
Exemplary Synthesis of Compound 52
Step 1
To a solution of EtOAc (4.42 g,50.19mmol,4.91mL,1 eq.) in THF (100 mL) was added dropwise a solution of LiHMDS (1 m,50.19mL,1 eq.) at-70 ℃ over a period of 30min under N2. Tert-butyl 4-oxopiperidine-1-carboxylate (10 g,50.19mmol,1 eq.) in THF (50 mL) was then added to the solution. The reaction mixture was stirred at-70℃for 1 hour. TLC (petroleum ether/ethyl acetate=10:1, rf=0.19) showed complete consumption of starting material. The residue was poured into water (100 mL). The aqueous phase was extracted with ethyl acetate (100 ml x 3). The combined organic phases were washed with brine (100 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (80 g,0% -40% (20 min) ethyl acetate/petroleum ether, 40% (10 min) ethyl acetate/petroleum ether) to give 4- (2-ethoxy-2-oxo-ethyl) -4-hydroxy-piperidine-1-carboxylic acid tert-butyl ester (11.2 g,38.98mmol,77.66% yield) as a colorless oil.
Step 2
To a mixture of tert-butyl 4- (2-ethoxy-2-oxo-ethyl) -4-hydroxy-piperidine-1-carboxylate (5 g,17.40mmol,1 eq.) in THF (50 mL) was added LiAlH4 (726.38 mg,19.14mmol,1.1 eq.) at 0 ℃ under N2. The mixture was then stirred at 25 ℃ for 1h to give a white suspension. TLC (petroleum ether: ethyl acetate=10:1, rf=0.10) showed no starting material and a new spot. The reaction mixture was quenched with water (1 mL), followed by the addition of 15% aqueous sodium hydroxide (1 mL) and water (3 mL). The solids were removed by filtration. The filtrate was concentrated under reduced pressure to give tert-butyl 4-hydroxy-4- (2-hydroxyethyl) piperidine-1-carboxylate (3.3 g,13.45mmol,77.31% yield) as a colorless oil.
Step 3
To a mixture of tert-butyl 4-hydroxy-4- (2-hydroxyethyl) piperidine-1-carboxylate (3.3 g,13.45mmol,1 eq.) and TosCl (3.85 g,20.18mmol,1.5 eq.) in DCM (30 mL) was added TEA (4.08 g,40.36mmol,5.62mL,3 eq.) at 25 ℃ in one portion. The mixture was stirred at 25℃for 16 hours. TLC (petroleum ether: ethyl acetate=10:1, rf=0.34, pma) showed no starting material and TLC showed new spots. LCMS showed the desired MS. The residue was poured into water (30 mL). The aqueous phase was extracted with ethyl acetate (50 ml x 3). The combined organic phases were washed with brine (50 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (40 g,0% -50% (10 min) ethyl acetate/petroleum ether, 50% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 4-hydroxy-4- [2- (p-toluenesulfonyloxy) ethyl ] piperidine-1-carboxylate (4.6 g,11.51mmol,85.60% yield) as a yellow oil.
Step 4
To a mixture of tert-butyl 4-hydroxy-4- [2- (p-toluenesulfonyloxy) ethyl ] piperidine-1-carboxylate (2 g,5.01mmol,1 eq.) and benzyl (3S) -3-methylpiperazine-1-carboxylate (3.52 g,15.02mmol,3 eq.) in MeCN (10 mL) was added KI (1.66 g,10.01mmol,2 eq.) and DIPEA (1.29 g,10.01mmol,1.74mL,2 eq.) at 20℃in one portion. The mixture was stirred at 80℃for 10 hours. LCMS showed the desired MS. The mixture was cooled to 25 ℃ and concentrated under reduced pressure at 25 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 33_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0; end B:35; flow rate: 25mL/min; gradient time: 40min;100% B hold time: 3 min) to give benzyl (3S) -4- [2- (1-tert-butoxycarbonyl-4-hydroxy-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylate (150 mg,298.96umol,5.97% yield, 92% purity) as a yellow gum.
Step 5
DAST (52.38 mg,324.96umol,42.93uL,1 eq.) was added dropwise to a mixture of benzyl (3S) -4- [2- (1-tert-butoxycarbonyl-4-hydroxy-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylate (150 mg,324.96umol,1 eq.) in DCM (10 mL) at-40 ℃. Then heated to 25 ℃ and stirred for 2 hours to give a colorless solution. TLC (petroleum ether: ethyl acetate=1:1, rf=0.13, pma) showed the reaction was complete and LCMS showed the desired MS. The residue was poured into NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with petroleum ether (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -100% (20 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [2- (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylate (122 mg,223.69umol,68.84% yield, 85% purity) as a yellow gum.
Step 6
To a mixture of benzyl (3S) -4- [2- (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylate (122 mg,263.17 mol,1 eq.) in DCM (5 mL) at 20 ℃ was added TFA (3.08 g,27.01mmol,2mL,102.65 eq.) in one portion. The mixture was stirred at 25℃for 30min. TLC showed the reaction was complete. The mixture was concentrated in vacuo to give (3S) -4- [2- (4-fluoro-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (150 mg,229.32umol,87.14% yield, 73% purity, TFA) as a yellow gum.
Step 7
To a mixture of (3S) -4- [2- (4-fluoro-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (150 mg,229.32umol,73% purity, 1 eq, TFA) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (84.71 mg,229.32umol,1 eq) in MeOH (10 mL) at 25 ℃ was added borane in one portion; 2-methylpyridine (49.06 mg,458.65umol,2 eq.) and HOAc (1 mL). The mixture was stirred at 25℃for 1h. TLC (dichloromethane: methanol=10:1, rf=0.43) showed the reaction was complete and LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10:1, rf= 0.43,0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give (3S) -4- [2- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] methyl ] -4-fluoro-4-piperidinyl ] ethyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (193 mg, crude) as a yellow oil.
Step 8
A mixture of (3S) -4- [2- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -4-fluoro-4-piperidinyl ] ethyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (193 mg,269.24umol,1 eq.) in TFA (2.78 g,24.39mmol,1.81mL,90.60 eq.) at 20℃in one portion. The mixture was stirred at 70 ℃ for 1h to give a yellow solution. TLC showed the reaction was complete. The residue was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [2- [ (2S) -2-methylpiperazin-1-yl ] ethyl ] -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione as a yellow gum (150 mg,131.36umol,48.79% yield, 71% purity, 2 TFA).
Step 9
DIEA (171.89 mg,1.33mmol,231.66uL,8 equiv.) was added in one portion to a mixture of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [ 4-fluoro-4- [2- [ (2S) -2-methylpiperazin-1-yl ] ethyl ] -1-piperidyl ] methyl ] -1-piperidyl ] isoindoline-1, 3-dione (150 mg,185.01 uL, 1.11 equiv., 2 TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (50 mg,166.26 uL, 1 equiv.) in DMSO (5 mL) at 20deg.C. The mixture was stirred at 80℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:40; flow: 25mL/min; gradient time: 40min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (16.9 mg,19.45 mol,11.70% yield, 97.5% purity) as a yellow solid.
Exemplary Synthesis of Compound 53
Step 1
A solution of tert-butyl 4- (2-bromoacetyl) piperidine-1-carboxylate (695.05 mg,2.27mmol,1 eq.) in MeCN (10 mL) was stirred at 20deg.C. Benzyl piperazine-1-carboxylate (500 mg,2.27mmol,438.60ul,1 eq.) was then added to the mixture and the mixture was heated at 25℃under N 2 Stirred for 2 hours. TLC (dichloromethane: methanol=10:1) showed complete consumption of starting material and found two new principal points. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% to 10% dichloromethane/methanol) to give 4- [2- (1-tert-butoxycarbonyl-4-piperidinyl) -2-oxo-ethyl ] as a yellow gum]Benzyl piperazine-1-carboxylate (69mg, 774.32umol,34.11% yield, 50% purity).
Step 2
4- [2- (1-tert-Butoxycarbonyl-4-piperidinyl) -2-oxo-ethyl]A solution of benzyl piperazine-1-carboxylate (69mg, 1.55mmol,1 eq.) in DCM (10 mL) was stirred at 0deg.C for 20min. DAST (8.74 g,54.20mmol,7.16mL,35 eq.) was then added to the mixture and the mixture was heated to 25℃under N 2 Stirred for 16 hours. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=1:1) showed complete consumption of starting material and found two new principal points. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% to 50% dichloromethane/methanol) to give the compound 4- [2- (1-tert-butoxycarbonyl-4-piperidinyl) -2, 2-difluoro-ethyl as a colourless gum ]Benzyl piperazine-1-carboxylate (298 mg,254.95umol,16.46% yield, 40% purity).
Step 3
To 4- [2- (1-tert-butoxycarbonyl-4-piperidinyl) -2, 2-difluoro-ethyl]To a solution of benzyl piperazine-1-carboxylate (298 mg,637.37umol,1 eq.) in DCM (3 mL) was added TFA (4.62 g,40.52mmol,3mL,63.57 eq.). The mixture was then brought to 25℃under N 2 Stirred for 0.5 hours. LCMS showed the desired MS. The residue was concentrated under reduced pressure to give 4- [2, 2-difluoro-2- (4-piperidinyl) ethyl ] as a brown oil]Piperazine-1-carboxylic acid benzyl ester (480 mg, crude, TFA).
Step 4
To 4- [2, 2-difluoro-2- (4-piperidinyl) ethyl group]Piperazine-1-carboxylic acid benzyl ester (380 mg,1.03mmol,1 eq), 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl]To a solution of piperidine-4-carbaldehyde (114.60 mg,310.26umol,0.3 eq.) in MeOH (10 mL) was added AcOH (2 mL), borane; 2-methylpyridine (221.24 mg,2.07mmol,2 eq.). The mixture was then brought to 20℃under N 2 Stirred for 2 hours. LCMS showed the desired MS. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% to 10% dichloromethane/methanol) to give 4- [2- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] as a yellow gum ]-4-piperidinyl]Methyl group]-4-piperidinyl]-2, 2-difluoro-ethyl group]Benzyl piperazine-1-carboxylate (1 g, crude material).
Step 5
To 4- [2- [1- [ [1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl ]]-4-piperidinyl]Methyl group]-4-piperidinyl]-2, 2-difluoro-ethyl group]To a mixture of benzyl piperazine-1-carboxylate (200 mg,277.47 uL,1 eq.) was added TFA (31.64 mg,277.47 uL, 20.54uL,1 eq.). The mixture is then brought to 70℃under N 2 Stirred for 0.5 hours. TLC (dichloromethane: methanol=10:1) showed complete consumption of starting material and found a new main point. The residue was concentrated under reduced pressure to give 5- [4- [ [4- (1, 1-difluoro-2-piperazin-1-yl-ethyl) -1-piperidinyl ] as a brown oil]Methyl group]-1-piperidinyl group]-2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (200 mg, crude, TFA).
Step 6
To 5- [4- [ [4- (1, 1-difluoro-2-piperazin-1-yl-ethyl) -1-piperidinyl]Methyl group]-1-piperidinyl group]To a solution of 2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (162 mg,276.13 mmole, 1 eq.) in DMSO (10 mL) was added 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (66.44 mg,220.91 mmole, 0.8 eq.) and DIEA (2.23 g,17.22 mmole, 3mL,62.37 eq.). The mixture is then brought to 70℃under N 2 Stirred for 16 hours. LCMS showed the desired MS. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -40%,40 min) to give 5- [4- [ [4- [1, 1-difluoro-2- [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a yellow solid]Pyrimidin-4-yl]Piperazin-1-yl]Ethyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (13.5 mg,15.23umol,5.52% yield, 96% purity).
Exemplary Synthesis of Compound 54
Compound 54 was prepared in a similar manner to compound 53 using benzyl 4- (2-piperazin-1-ylethyl) piperazine-1-carboxylate.
Step 1
At 25 ℃ at N 2 Downward 4- (2-chloroethyl) piperazine-1-carboxylic acid tert-butyl esterEsters (1 g,4.02mmol,1 eq.) and benzyl piperazine-1-carboxylate (1.33 g,6.03mmol,1.17mL,1.5 eq.) in EtOH (10 mL) and H 2 To the mixture in O (1 mL) was added NaHCO in one portion 3 (675.43 mg,8.04mmol,312.70uL,2 eq.). The mixture was stirred at 80℃for 3 hours. LCMS showed the desired MS. The mixture was cooled to 25 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (20 g,0% -40% (10 min) ethyl acetate/petroleum ether, 40% (10 min) ethyl acetate/petroleum ether) to give 4- [2- (4-tert-butoxycarbonylpiperazin-1-yl) ethyl as a yellow solid]Benzyl piperazine-1-carboxylate (800 mg,1.85mmol,46.01% yield).
Step 2
To a mixture of benzyl 4- [2- (4-tert-butoxycarbonylpiperazin-1-yl) ethyl ] piperazine-1-carboxylate (200 mg,462.37umol,1 eq.) in DCM (5 mL) was added TFA (1.85 g,16.25mmol,1.20mL,35.15 eq.) at 25℃in one portion. The mixture was stirred at 25℃for 30min. LCMS (EB 16-1124-P1 A1) showed the reaction was complete. The residue was concentrated in vacuo to give benzyl 4- (2-piperazin-1-ylethyl) piperazine-1-carboxylate as a yellow gum (200 mg,277.74umol,60.07% yield, 62% purity, TFA).
Exemplary Synthesis of Compound 55
Compound 55 was prepared in a similar manner to compound 54 starting from tert-butyl (2S) -2-methylpiperazine-1-carboxylate.
Exemplary Synthesis of Compound 56
Step 1
To a solution of tert-butyl piperidine-4-carboxylate (503.03 mg,2.72mmol,1.5 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (500 mg,1.81mmol,1 eq.) in DMSO (5 mL) and DIEA (2.34 g,18.10mmol,3.15mL,10 eq.). The mixture was stirred at 80℃for 16h. LCMS showed the desired product MS. The resulting product was poured into H2O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (0% -50% (10 min) ethyl acetate/petroleum ether, 50% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carboxylate (660 mg,1.49mmol,82.59% yield) as a yellow solid.
Step 2
To a solution of tert-butyl 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carboxylate (660 mg,1.49mmol,1 eq.) in DCM (5 mL) and TFA (3.08 g,27.01mmol,2mL,18.07 eq.). The mixture was stirred at 25℃for 30min. LCMS showed the desired product MS. The resulting product was concentrated in vacuo to give a residue. The residue was purified by preparative HPLC (column: xtime C18:40 mm 10um; mobile phase: [ water (0.05% HCl) -ACN ]; B%:15% -45%,10 min) to give 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] piperidine-4-carboxylic acid (550 mg,1.40mmol,93.49% yield, 97.93% purity) as a yellow solid.
Step 3
To a solution of benzyl (3S) -3-methyl-4- (2-piperazin-1-ylethyl) piperazine-1-carboxylate (130 mg,339.49umol,1.31 eq, HCl) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carboxylate (100 mg,259.49umol,1 eq) in DMF (3 mL) was added DIEA (100.61 mg,778.47umol,135.60ul,3 eq) and HATU (98.67 mg,259.49umol,1 eq). After the addition, the reaction mixture was stirred at 20 ℃ for 1h. LCMS and TLC (dichloromethane: methanol=10:1) showed the reaction was complete. The reaction mixture was diluted with ethyl acetate (20 mL) and washed with brine (10 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 15% methanol in dichloromethane) to give (3S) -4- [2- [4- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbonyl ] piperazin-1-yl ] ethyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (95 mg,101.01umol,38.93% yield, 75.9% purity) as a yellow solid.
Step 4
A mixture of (3S) -4- [2- [4- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbonyl ] piperazin-1-yl ] ethyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (95 mg,133.09umol,1 eq.) and TFA (3.08 g,27.01mmol,2mL,202.97 eq.) was stirred at 80℃for 1h. LCMS (EB 12-916-P1B) showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- [2- [ (2S) -2-methylpiperazin-1-yl ] ethyl ] piperazine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (80 mg, crude) as a yellow gum.
Step 5
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- [2- [ (2S) -2-methylpiperazin-1-yl ] ethyl ] piperazine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (67.46 mg,116.38umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (35 mg,116.38umol,1 eq.) in DMSO (2 mL) was added DIEA (75.21 mg,581.89umol,101.36uL,5 eq.). After the addition, the reaction solution was stirred at 90 ℃ for 16h. LCMS showed the reaction was complete. The reaction mixture was diluted with brine (10 mL) and extracted with dichloromethane (20 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] piperazine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (15.2 mg,17.53umol,15.07% yield, 97.35% purity) as a yellow solid.
Exemplary Synthesis of Compound 57
Compound 57 was prepared in a similar manner to compound 56 using benzyl (3S) -3-methyl-4- [2- (4-piperidinyl) ethyl ] piperazine-1-carboxylate.
Step 1
To a solution of tert-butyl 4- (2-oxoethyl) piperidine-1-carboxylate (291.04 mg,1.28mmol,1 eq.) and benzyl (3S) -3-methylpiperazine-1-carboxylate (300 mg,1.28mmol,1 eq.) in MeOH (5 mL) and AcOH (0.5 mL) was added borane; 2-methylpyridine (273.92 mg,2.56mmol,2 eq.). After the addition, the mixture was stirred at 50 ℃ for 3 hours. LCMS showed the desired MS. The residue was poured into NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,100-200 mesh silica gel, 0% -100% (15 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [2- (1-tert-butoxycarbonyl-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylate (500 mg,1.12mmol,87.63% yield) as a colorless oil.
Step 2
To a mixture of benzyl (3S) -4- [2- (1-tert-butoxycarbonyl-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylate (100 mg,224.42umol,1 eq.) in EtOAc (2 mL) was added HCl/EtOAc (2M, 2mL,17.82 eq.). The mixture was stirred at 20℃for 1h. TLC showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give benzyl (3S) -3-methyl-4- [2- (4-piperidinyl) ethyl ] piperazine-1-carboxylate (77 mg, crude, HCl) as a white solid.
Exemplary Synthesis of Compound 58
Compound 58 was prepared in a similar manner to compound 56 using 1- [2- [ (2S) -4-benzyloxycarbonyl-2-methyl-piperazin-1-yl ] ethyl ] piperidine-4-carboxylic acid and 2- (2, 6-dioxo-3-piperidinyl) -5-piperazin-1-yl-isoindoline-1, 3-dione.
Step 1
(3S) -4- (2-chloroethyl) -3-methyl-piperazine-1-carboxylic acid benzyl ester (1.23 g,4.16mmol,1.1 eq.) and piperidine-4-carboxylic acid tert-butyl ester (700 mg,3.78mmol,1 eq.) were dissolvedSolution in EtOH (5 mL) and H2O (0.5 mL) followed by addition of NaHCO3 (952.24 mg,11.34mmol,440.85uL,3 eq.) to the reaction and stirring at 80℃for 4H. LCMS showed about 60% of the desired compound and TLC (dichloromethane: methanol=10:1, rf=0.39, I2) showed formation of the main spot. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel20g/>Silica gel flash column, eluent: 0% -4% methylene chloride methanol, 40 mL/min). The compound (3S) -4- [2- (4-tert-butoxycarbonyl-1-piperidinyl) ethyl was obtained as a pale yellow gum]-3-methyl-piperazine-1-carboxylic acid benzyl ester (1.24 g,2.78mmol,73.65% yield).
Step 2
To a solution of benzyl (3S) -4- [2- (4-tert-butoxycarbonyl-1-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylate (300 mg,673.26umol,1 eq) in DCM (5 mL) was added TFA (3.46 g,30.39mmol,2.25mL,45.14 eq), followed by stirring the mixture at 25 ℃ for 4 hours. LCMS showed the desired compound and no starting material remained. The reaction mixture was concentrated under reduced pressure to give a residue. The compound 1- [2- [ (2S) -4-benzyloxycarbonyl-2-methyl-piperazin-1-yl ] ethyl ] piperidine-4-carboxylic acid (300 mg, crude) was obtained as a pale yellow oil.
Exemplary Synthesis of Compound 59
Step 1
A mixture of 4-piperidylmethanol (1 g,8.68mmol,1 eq), 2-chloroacetyl chloride (1.18 g,10.42mmol,828.71uL,1.2 eq) and TEA (1.82 g,17.96mmol,2.50mL,2.07 eq) in DCM (20 mL) was degassed and purged 3 times with N2, then the mixture was stirred at 0deg.C under an atmosphere of N2 for 3 hours. TLC indicated no residue of reactant 1 and a major new spot of lower polarity was detected. An aqueous HC1 solution (0.5M, 30 mL) was added to the reaction mixture. Will beThe reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (15 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel20g/>Silica gel flash column, eluent: 0% -100% ethyl acetate/petroleum ether gradient, 45 mL/min) to give 2-chloro-1- [4- (hydroxymethyl) -1-piperidinyl as a pale yellow oil]Ethanone (479 mg,1.72mmol,19.86% yield, 69% purity).
Step 2
To a solution of 2-chloro-1- [4- (hydroxymethyl) -1-piperidinyl ] ethanone (100 mg,521.77 mol,1.1 eq.) and benzyl (3S) -3-methylpiperazine-1-carboxylate (111.13 mg,474.34 mol,1 eq.) in MeCN (2 mL) was added DIEA (306.52 mg,2.37mmol,413.10ul,5 eq.) at 25 ℃. The mixture was stirred at 60℃for 16 hours. LC-MS (EB 2049-138-P1A) showed complete consumption of reactant 1 and the required mass was detected. The mixture was cooled to room temperature and concentrated, and the residue was extracted with ethyl acetate (10 mL). The organic layer was washed with water (10 ml×2), brine (10 ml×2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give benzyl (3S) -4- [2- [4- (hydroxymethyl) -1-piperidinyl ] -2-oxo-ethyl ] -3-methyl-piperazine-1-carboxylate (84 mg,109.99umol,23.19% yield, 51% purity) as a pale yellow oil. The crude product was used in the next step without further purification.
Step 3
To (3S) -4- [2- [4- (hydroxymethyl) -1-piperidinyl]-2-oxo-ethyl]To a solution of tert-butyl 3-methyl-piperazine-1-carboxylate (500 mg,1.41mmol,1 eq.) in DCM (5 mL) was added DMP (1.19 g,2.81mmol,870.93uL,2 eq.). The mixture was stirred at 25℃for 2 hours. LC-MS (EB 2049-145-P1A) showed complete consumption of reactant 1 and the required mass was detected. The reaction mixture was diluted with water (10 mL)And extracted with ethyl acetate (10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel20/>Silica gel flash column, eluent: 0% -100% ethyl acetate/Petroleum ether gradient, 40 mL/min) to give (3S) -4- [2- (4-formyl-1-piperidinyl) -2-oxo-ethyl as a colorless oil]-3-methyl-piperazine-1-carboxylic acid tert-butyl ester (240 mg,679.01umol,48.27% yield).
Step 4
To (3S) -4- [2- (4-formyl-1-piperidinyl) -2-oxo-ethyl]To a solution of tert-butyl 3-methyl-piperazine-1-carboxylate (110 mg,311.21umol,1 eq) and 2- (2, 6-dioxo-3-piperidinyl) -5-piperazin-1-yl-isoindoline-1, 3-dione (127.85 mg,373.46umol,1.2 eq) in MeOH (5 mL) and HOAc (1 mL) was added borane; 2-methylpyridine (66.58 mg, 622.43. Mu.l, 2 eq.) and DIEA (80.44 mg, 622.43. Mu.l, 108.42. Mu.l, 2 eq.). The mixture was stirred at 25℃for 16 hours. LC-MS (EB 2049-148-P1A) showed complete consumption of reactant 1 and the required mass was detected. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel 12g/>Silica gel flash column, eluent: 0% -100% (20 min) ethyl acetate/petroleum ether gradient, 40mL/min; purification with 0% -10% (15 min) MeOH/DCM,35 mL/min) gave (3S) -4- [2- [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] as a pale yellow oil]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-2-oxo-ethyl]-3-methyl-piperazine-1-carboxylic acid tert-butyl ester (200 mg,294.20umol,94.53% yield).
Step 5
To a solution of (3S) -4- [2- [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -2-oxo-ethyl ] -3-methyl-piperazine-1-carboxylic acid tert-butyl ester (200 mg,294.20umol,1 eq.) in DCM (5 mL) was added TFA (5 mL). The mixture was stirred at 25℃for 1 hour. TLC indicated no residue of reactant 1 and a major new spot of greater polarity was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [2- [ (2S) -2-methylpiperazin-1-yl ] acetyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (560 mg, crude, 12 TFA) as a pale yellow oil. The crude product was used in the next step without further purification.
Step 6
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [2- [ (2S) -2-methylpiperazin-1-yl ] acetyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (560 mg,287.48umol,1 eq, 12 TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (35 mg,116.38umol,4.05e-1 eq) in DMSO (3 mL) was added DIEA (445.86 mg,3.45mmol,600.88uL,12 eq). The mixture was stirred at 110℃for 16 hours. LC-MS (EB 2049-152-P1A) showed complete consumption of reactant 1 and the required mass was detected. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by reverse phase HPLC (column Phenomenex Luna C18 75 x 30mm x 3um; mobile phase: [ water (0.225% FA) -ACN;: 18% -48%,35 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] acetyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (41 mg,48.58umol,16.90% yield, 100% purity) as a yellow solid.
Exemplary Synthesis of Compound 60
Step 1
To a solution of ethyl 4-hydroxycyclohexane carboxylate (1 g,5.81mmol,1 eq.) in THF (15 mL) at 0deg.C were added TEA (881.33 mg,8.71mmol,1.21mL,1.5 eq.) and TMSCL (693.91 mg,6.39mmol,810.65uL,1.1 eq.) and the reaction mixture was stirred at 25deg.C for 1 hour. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. To a stirred solution of the above residue and benzyl 4-formylpiperidine-1-carboxylate (1.72 g,6.97mmol,1.2 eq.) in DCM (15 mL) was then added Et3SiH (810.19 mg,6.97mmol,1.11mL,1.2 eq.) and TMSOTF (709.80 mg,3.19mmol,577.07uL,0.55 eq.) dropwise at-60℃and the reaction mixture stirred at 0℃under N2 for 2 hours. TLC (petroleum ether: ethyl acetate=3:1, pma, rf=0.52) showed formation of a main spot. The reaction mixture was quenched by addition of water (30 mL) and extracted with DCM (30 mL x 3). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel40g/>Silica gel flash column, eluent: 0% -17% ethyl acetate/petroleum ether gradient, 50 mL/min). Obtaining the compound 4- [ (4-ethoxycarbonylcyclohexyloxy) methyl as colorless oil ]Benzyl piperidine-1-carboxylate (1.88 g,4.66mmol,80.24% yield).
Step 2
DIBALH (1M, 1.36mL,1.1 eq.) was added dropwise to a solution of benzyl 4- [ (4-ethoxycarbonylcyclohexyloxy) methyl ] piperidine-1-carboxylate (500 mg,1.24mmol,1 eq.) in DCM (10 mL) at-70℃over a period of 30min under N2. During which the temperature is kept below-70 ℃. The reaction mixture was warmed to 0 ℃ and stirred for 1.5 hours. LCMS showed about 71% of the desired compound and the starting material was completely consumed. Saturated NH4Cl was added to the reaction mixture under ice-cooling. Saturated brine was added thereto, followed by extraction with DCM (80 ml×3). The organic layer was dried over anhydrous magnesium sulfate and concentrated. Compound 4- [ [4- (hydroxymethyl) cyclohexyloxy ] methyl ] piperidine-1-carboxylic acid benzyl ester
Step 3
At 0 ℃ to 4- [ [4- (hydroxymethyl) cyclohexyloxy ]]Methyl group]To a mixture of benzyl piperidine-1-carboxylate (440 mg,1.22mmol,1 eq), DMAP (44.61 mg,365.17 mmole, 0.3 eq.) and TEA (369.51 mg,3.65mmol,508.27uL,3 eq.) in DCM (10 mL) was added TosCl (348.09 mg,1.83mmol,1.5 eq.). The mixture was stirred at 25℃for 4 hours. LCMS showed about 36% of the desired compound and TLC (petroleum ether: ethyl acetate=1:1, rf=0.57, pma) showed formation of a new main spot. The reaction mixture was quenched by addition of water (30 mL) and extracted with DCM (30 mL x 3). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel 20g/>Silica gel flash column, eluent: 0% -38% ethyl acetate/petroleum ether gradient, 40 mL/min). Obtaining the compound 4- [ [4- (p-toluenesulfonyloxy methyl) cyclohexyloxy ] as a colorless oil]Methyl group]Benzyl piperidine-1-carboxylate (300 mg,523.60umol,43.02% yield, 90% purity).
Step 4
To 4- [ [4- (p-toluenesulfonyloxymethyl) cyclohexyloxy ]]Methyl group]Benzyl piperidine-1-carboxylate (300 mg,581.78umol,1 eq) and 2- (2, 6-dioxo-3-piperidinyl) -5-piperazin-1-yl-isoindoline-1, 3-dione (239.01 mg,698.13umol,1.2 eq) in CH3CN (10 mL) was added KI (965.76 mg,5.82mmol,10 eq) and DIEA (751.91 mg,5.82mmol,1.01mL,10 eq), then the mixture was heated to 100deg.CStirring for 16 hours. LCMS showed the desired compound and complete consumption of starting material, TLC (methanol: dichloromethane=10:1, rf=0.43, uv=254 nm) showed formation of new spots. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel12g/>Silica gel flash column, eluent: 0% -3% methanol in dichloromethane, 40 mL/min). Obtaining the compound 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl ] as a yellow solid ]Piperazin-1-yl]Methyl group]Cyclohexyloxy radical]Methyl group]Benzyl piperidine-1-carboxylate (760 mg,554.09umol,95.24% yield, 50% purity).
Step 5
To a solution of benzyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] cyclohexyloxy ] methyl ] piperidine-1-carboxylate (300 mg,437.44umol,1 eq.) in TFA (6.93 g,60.78mmol,4.50ml,138.94 eq.) and stirring the solution at 70 ℃ for 40min. LCMS showed about 66% of the desired compound and the starting material was completely consumed. The reaction mixture was concentrated under reduced pressure to give a residue. The compound 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (4-piperidinylmethoxy) cyclohexyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (310 mg, crude, TFA) was obtained as a pale yellow oil.
Step 6
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (4-piperidinylmethoxy) cyclohexyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (50 mg,90.63umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (35.43 mg,117.82umol,1.3 eq.) in DMSO (3 mL) was added DIEA (117.14 mg,906.33umol,157.87ul,10 eq.) and the mixture was stirred at 90 ℃ for 16 hours. LCMS showed about 42.8% of the desired compound and starting material was consumed. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min). The compound 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methoxy ] cyclohexyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (16.5 mg,19.98umol,22.04% yield, 98.8% purity) was obtained as a yellow solid.
Exemplary Synthesis of Compound 61
Step 1
A mixture of tert-butyl 4- (hydroxymethyl) piperidine-1-carboxylate (1 g,4.64mmol,1 eq.) and MsCl (585.29 mg,5.11mmol,395.46uL,1.1 eq.) in DCM (20 mL) was treated dropwise with TEA (940.03 mg,9.29mmol,1.29mL,2 eq.) at 0deg.C. The reaction mixture was stirred at 0℃for 1.5h. TLC (petroleum ether: ethyl acetate=5:1, rf=0.53) showed no starting material and a new spot. The mixture was diluted with DCM (25 mL x 3) and washed with 1m NaHCO3 (200 mL x 2) and brine (25 mL). The DCM layer was dried over Na2SO4, filtered, and the filtrate was concentrated to give a colorless oil.
Step 2
To a solution of benzyl 4-hydroxypiperidine-1-carboxylate (1.13 g,4.81mmol,1 eq.) in DMF (12 mL) was added NaH (433.33 mg,10.83mmol,60% purity, 2.25 eq.) at 0deg.C. The mixture was stirred at 0℃for 0.5 h. After this time, tert-butyl 4- (methylsulfonylmethyl) piperidine-1-carboxylate (1.41 g,4.81mmol,1 eq.) was added and the mixture stirred at 60℃for 16h. LCMS showed the desired MS and no starting material. After completion, the reaction mixture was quenched with water (30 mL) at 0 ℃ and extracted with EtOAc (15 mL x 3). The combined organic layers were dried over Na2SO4, filtered and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10:1) to give tert-butyl 4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxymethyl ] piperidine-1-carboxylate (277 mg,640.39umol,13.31% yield) as a colorless oil.
Step (a)3
To a solution of tert-butyl 4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxymethyl ] piperidine-1-carboxylate (270 mg,624.20umol,1 eq.) in EtOH (5 mL) was added Pd/C (30 mg,25.45umol,10% purity, 4.08e-2 eq.) under N2. The suspension was degassed under vacuum and purged three times with H2. The mixture was stirred at 25℃under H2 (15 psi) for 1H. LCMS showed the desired MS and no starting material. The reaction mixture was concentrated in vacuo to give the crude product as a colorless oil.
Step 4
To a solution of tert-butyl 4- (4-piperidinyloxymethyl) piperidine-1-carboxylate (180 mg,603.18umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (222.80 mg,603.18umol,1 eq) in MeOH (10 mL) was added HOAc (1 mL) and stirred at 25 ℃ for 10min. Then adding borane; 2-methylpyridine (129.03 mg,1.21mmol,2 eq.) and the mixture was stirred at 25℃under N2 for 16h. LCMS showed the reaction was complete. To the residue was added water (5 mL). The aqueous phase was extracted with ethyl acetate (8 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by Combi Flash (0% -8% MeOH/DCM) to give tert-butyl 4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -4-piperidinyl ] oxymethyl ] piperidine-1-carboxylate (390 mg,598.35umol,99.20% yield) as a green oil.
Step 5
To a mixture of tert-butyl 4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -4-piperidinyl ] oxymethyl ] piperidine-1-carboxylate (120 mg,184.11umol,1 eq) in DCM (4 mL) was added TFA (1.54 g,13.51mmol,1mL,73.36 eq) at 25℃followed by stirring for 1 hour. LCMS showed 91% of the desired compound and no starting material. The mixture was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (4-piperidinylmethoxy) -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (105 mg, crude, TFA) as a yellow oil.
Step 6
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (4-piperidinylmethoxy) -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (100 mg,150.22umol,1 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (24.86 mg,82.67umol,0.55 eq) in DMSO (3 mL) was added DIEA (232.97 mg,1.80mmol,313.98ul,12 eq) and stirred at 90 ℃ for 16 hours. LCMS showed 64% of the desired compound and no starting material. The mixture was poured into water (15 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -40%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methoxy ] -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (30.1 mg,35.41umol,23.57% yield, 96% purity) as a yellow solid.
Exemplary Synthesis of Compound 62
Step 1
To a mixture of 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carboxylic acid (103.31 mg,268.08umol,1 eq.) in DMF (6 mL) was added HATU (132.51 mg,348.50umol,1.3 eq.) followed by stirring the mixture at 25℃for 30min, followed by the addition of tert-butyl 4- (4-piperidinyloxymethyl) piperidine-1-carboxylate (80 mg,268.08umol,1 eq.) and DIEA (103.94 mg,804.24umol,140.08uL,3 eq.) and stirring the mixture for 1h. TLC (DCM: meoh=10:1, rf=0.58) showed no starting material and a new spot. To the residue was added water (5 mL). The aqueous phase was extracted with ethyl acetate (8 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. This was purified by Combi Flash (SiO 2 11% DCM/MeOH) to give tert-butyl 4- [ [1- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbonyl ] -4-piperidinyl ] oxymethyl ] piperidine-1-carboxylate (150 mg,225.30umol,84.04% yield) as a green oil.
Step 2
To a mixture of tert-butyl 4- [ [1- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbonyl ] -4-piperidinyl ] oxymethyl ] piperidine-1-carboxylate (150 mg,225.30umol,1 eq.) in DCM (6 mL) was added TFA (2.31 g,20.26mmol,1.5mL,89.92 eq.) followed by stirring for 1h. TLC (petroleum ether/ethyl acetate=1:1) showed no starting material and new spots. The mixture was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- (4-piperidinylmethoxy) piperidine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (150 mg, crude, TFA) as a yellow oil.
Step 3
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- (4-piperidinylmethoxy) piperidine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (150 mg,220.69umol,1 eq., TFA), 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (40 mg,133.00umol,6.03e-1 eq.) in DMSO (4 mL) was added DIEA (342.27 mg,2.65mmol,461.29uL,12 eq.) and stirred at 90℃for 16H. LCMS showed the reaction was complete. The residue was concentrated under reduced pressure to give a residue. This was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -40%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methoxy ] piperidine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (14.3 mg,16.89umol,7.65% yield, 98% purity) as a yellow solid.
Exemplary Synthesis of Compound 63
Step 1
To 1-imino-1-oxo-1, 4-thiazine-4-carboxylic acid tert-butyl ester (100 mg,426.78 mol,1 eq.), 4-formylpiperidine-1-carboxylic acid benzyl ester (158.31 mg,640.16 mol,1.5 eq.), borane; to a mixture of 2-methylpyridine (136.95 mg,1.28mmol,3 eq.) in MeOH (10 mL) was added AcOH (2 mL) borane; 2-methylpyridine (136.95 mg,1.28mmol,3 eq.) then the mixture was stirred at 25℃under an atmosphere of N2 for 16 hours. LCMS showed complete consumption of starting material and found the desired MS. TLC (petroleum ether: ethyl acetate=0:1) showed several new spots. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 80% ethyl acetate/petroleum ether) to give tert-butyl 1- [ (1-benzyloxycarbonyl-4-piperidinyl) methylimino ] -1-oxo-1, 4-thiazine-4-carboxylate (199mg, 260.71umol,61.09% yield, 61% purity) as a colorless gum.
Step 2
To a mixture of tert-butyl 1- [ (1-benzyloxycarbonyl-4-piperidinyl) methylimino ] -1-oxo-1, 4-thiazine-4-carboxylate (199mg, 427.40 mol,1 eq) in DCM (3 mL) was added TFA (4.62 g,40.52mmol,3mL,94.80 eq), and the mixture was stirred at 25℃for 1 h. TLC (petroleum ether: ethyl acetate=0:1) showed a new major spot. The reaction mixture was concentrated under reduced pressure to give benzyl 4- [ [ (1-oxo-1, 4-thiazinan-1-ylidene) amino ] methyl ] piperidine-1-carboxylate (200 mg, crude, TFA) as a yellow gum.
Step 3
To a mixture of benzyl 4- [ [ (1-oxo-1, 4-thiazinan-1-ylidene) amino ] methyl ] piperidine-1-carboxylate (156 mg,426.83umol,1 eq), 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (157.66 mg,426.83umol,1 eq) in MeOH (10 mL) was added AcOH (1 mL), borane; 2-methylpyridine (91.31 mg,853.65umol,2 eq.) the mixture was then stirred at 25℃under an atmosphere of N2 for 2 hours. LCMS showed complete consumption of starting material and found the desired MS. TLC (dichloromethane: methanol=10:1) showed several new spots. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 30% methanol/dichloromethane) to give benzyl 4- [ [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -1-oxo-1, 4-thiazinan-1-ylidene ] amino ] methyl ] piperidine-1-carboxylate (200 mg,180.84umol,42.37% yield, 65% purity) as a yellow gum.
Step 4
To a mixture of 4- [ [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -1-oxo-1, 4-thiazinan-1-ylidene ] amino ] methyl ] piperidine-1-carboxylic acid benzyl ester (200 mg,278.22umol,1 eq.) in TFA (31.72 mg,278.22umol,20.60ul,1 eq.) the mixture was then stirred at 70 ℃ under an atmosphere of N2 for 1 hour. TLC (dichloromethane: methanol=10:1) showed a new spot. The reaction mixture was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 1-oxo-1- (4-piperidinylmethylimino) -1, 4-thiazinan-4-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (200 mg, crude, TFA) as a yellow gum.
Step 5
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 1-oxo-1- (4-piperidinylmethylimino) -1, 4-thiazinan-4-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (58.33 mg,99.75umol,1 eq.) 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (30 mg,99.75umol,1 eq.) in DMSO (3 mL) was added DIEA (38.68 mg,299.26umol,52.13uL,3 eq.) and the mixture was stirred at 100℃under N2 atmosphere for 12 hours. LCMS showed complete consumption of starting material and found the desired MS. The reaction mixture was filtered and concentrated under reduced pressure. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methylimino ] -1-oxo-1, 4-thiazinan-4-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (25.3 mg,29.50umol,29.57% yield, 99% purity) as a yellow solid.
Exemplary Synthesis of Compound 64
Step 1
To a solution of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (600 mg,1.86mmol,1 eq.) and 4- (dimethoxymethyl) piperidine (739.11 mg,4.64mmol,2.5 eq.) in DMSO (10 mL) was added Pd-PEPSI-pent Cl-o-methylpyridine (100.82 mg,185.68umol,0.1 eq.) and Cs2CO3 (1.21 g,3.71mmol,2 eq.) and stirred at 80℃under N2 for 16h. TLC (petroleum ether: ethyl acetate=0:1, rf=0.4) showed a new spot for the reaction. The reaction was quenched with NH4Cl (20 mL) and extracted with ethyl acetate (3 x 20 mL). The combined organic phases were washed with water, dried over Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 1/100) to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (560 mg,1.39mmol,75.13% yield) as a yellow solid.
Step 2
A solution of 3- [5- [4- (dimethoxymethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (70 mg,174.37umol,1 eq.) in THF (2 mL) and HCl (2M, 2mL,22.94 eq.) was stirred at 20℃for 2 h. The reaction mixture was poured into H2O (20 mL) and basified with aqueous NaHCO3 to ph=8. The mixture was extracted with ethyl acetate (20 ml x 5) and dried over anhydrous Na2SO4, concentrated in vacuo to give a residue. The residue was used directly in the next step without any purification. The compound 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (61 mg,159.63umol,91.55% yield, 93% purity) was a yellow solid.
Step 3
To a solution of benzyl (3S) -4- [2- (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) ethyl ] -3-methyl-piperazine-1-carboxylate (410 mg,884.41umol,1 eq.) in EtOH (20 mL) was added Pd/C (500 mg,1.77mmol,197.63uL,10% purity, 2 eq.) under N2. The suspension was degassed under vacuum and purged several times with H2. The mixture was stirred at 25℃under H2 (15 psi) for 1H. TLC showed the reaction was complete. The suspension was filtered through celite or a pad of silica gel and the filter cake was washed with ethyl acetate (50 ml×3). The combined filtrates were concentrated in vacuo to give tert-butyl 4-fluoro-4- [2- [ (2S) -2-methylpiperazin-1-yl ] ethyl ] piperidine-1-carboxylate (270 mg, crude) as a yellow gum.
Step 4
To a mixture of 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (250 mg,580.04umol,1 eq.) and tert-butyl 4-fluoro-4- [2- [ (2S) -2-methylpiperazin-1-yl ] ethyl ] piperidine-1-carboxylate (270 mg,819.54umol,1.41 eq.) in DMSO (5 mL) was added Et3N (176.08 mg,1.74mmol,242.21ul,3 eq.) at once followed by stirring at 100 ℃ for 1h. TLC and LCMS showed complete consumption of starting material. The mixture was cooled to 20 ℃, then the residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (3X 5 mL). The combined organic phases were washed with brine (2×5 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -5% (5 min) ethyl acetate/petroleum ether, 5% (10 min) ethyl acetate/petroleum ether, 5% -30% (5 min) ethyl acetate/petroleum ether, 30% (5 min) ethyl acetate/petroleum ether). The crude material was purified by preparative TLC (petroleum ether: ethyl acetate=10:1) to give 4-fluoro-4- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] piperidine-1-carboxylic acid tert-butyl ester (140 mg,190.59umol,32.86% yield, 98.56% purity) as a yellow oil.
Step 5
To a mixture of tert-butyl 4-fluoro-4- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] piperidine-1-carboxylate (140 mg,193.37umol,1 eq) in MeOH (5 mL) was added HCl/EtOAc (4 m,2mL,41.37 eq) in one portion at 25 ℃. The mixture was stirred at 65℃for 0.5h. LCMS showed the reaction was complete. The mixture was concentrated in vacuo to give 3- [6- [ (3S) -4- [2- (4-fluoro-4-piperidinyl) ethyl ] -3-methyl-piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (100 mg,181.63umol,93.93% yield, 96.28% purity, HCl) as a yellow solid.
Step 6
To a mixture of 3- [6- [ (3S) -4- [2- (4-fluoro-4-piperidinyl) ethyl ] -3-methyl-piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (100 mg,202.59 mol,1.33 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (54 mg,151.95 mol,1 eq) in MeOH (10 mL) at 20 ℃ was added borane in one portion; 2-methylpyridine (32.50 mg, 303.89. Mu. Mol,2 eq.) and HOAc (1 mL). The mixture was stirred at 20℃for 16h. LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,40 min) to afford 3- [5- [4- [ [ 4-fluoro-4- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] -1-piperidinyl ] methyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (22.5 mg,26.69umol,17.57% yield, 98.82% purity) as an off-white solid.
Exemplary Synthesis of Compound 65
Compound 65 was prepared in a similar manner to compound 64.
Exemplary Synthesis of Compound 66
Compound 66 was prepared in a similar manner to compound 64.
Exemplary Synthesis of Compound 67
Step 1
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (4-piperidinylmethyl) piperazin-1-yl ] isoindoline-1, 3-dione (300 mg,682.58 mol,1 eq, TFA) and tert-butyl 4-formylpiperidine-1-carboxylate (218.36 mg,1.02mmol,1.5 eq) in MeOH (5 mL) at 20 ℃ was added HOAc (0.5 mL) and borane in one portion; 2-methylpyridine (146.02 mg,1.37mmol,2 eq.). The solution was stirred at 20℃for 16h. TLC (DCM: meoh=10:1, rf=0.43) showed that the reaction was complete and the main spot (rf=0.43) was shown on TLC. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (3X 20 mL). The combined organic phases were washed with brine (2×20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (column: 12g,100-200 mesh silica gel, 0% -10% (5 min) MeOH/DCM,10% (10 min) MeOH/DCM) to give tert-butyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (430 mg,675.27umol,98.93% yield) as a yellow gum.
Step 2
To a solution of tert-butyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (430 mg,675.27umol,1 eq.) in DCM (2 mL) at 20℃was added TFA (230.98 mg,2.03mmol,149.99uL,3 eq.) in one portion. The mixture was stirred at 20℃for 30min to give a yellow solution. TLC (DCM: meoh=10:1, rf=0.43) showed completion of starting material. The solution was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (500 mg,621.15umol,91.99% yield, 95% purity, 2 TFA) as a yellow gum.
Step 3
To a mixture of 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (60 mg,199.51umol,1 eq) and 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (107.07 mg,199.51umol,1 eq) in DMSO (5 mL) was added DIEA (77.35 mg,598.52umol,104.25ul,3 eq) at 20 ℃ in one portion. The mixture was stirred at 80℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:30; flow rate: 25mL/min; gradient time: 35min;100% B hold time: 1 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (15.6 mg,19.36umol,9.70% yield, 99.39% purity) as a yellow solid.
Exemplary Synthesis of Compound 68
Step 1
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindoline-1, 3-dione (325 mg,587.14umol,1 eq. TFA) in DCM (5 mL) was added DIEA (379.42 mg,2.94mmol,511.35uL,5 eq.) and stirred at 20℃for 10min. The mixture was then concentrated. A solution of the residue and tert-butyl 4-formylpiperidine-1-carboxylate (125.22 mg,587.14umol,1 eq.) in HOAC (1 mL) and MeOH (10 mL) was stirred at 20deg.C for 20min, followed by the addition of borane; 2-methylpyridine (125.60 mg,1.17mmol,2 eq.). The mixture was then stirred at 25 ℃ under N2 for 16h. TLC (dichloromethane: methanol=10:1, rf=0.3) showed no starting material and a new spot. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% to 20% dichloromethane/methanol) to give tert-butyl 4- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (460 mg,570.68umol,97.20% yield, 79% purity) as a yellow solid.
Step 2
To a solution of tert-butyl 4- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (460 mg,722.38umol,1 eq.) in DCM (3 mL) was then added TFA (3.08 g,27.01mmol,2mL,37.39 eq.). The mixture was then stirred at 20℃for 1h. TLC (dichloromethane: methanol=10:1, rf=0.01) showed no starting material and a new spot. The residue was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (4-piperidinylmethyl) piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (387 mg, crude, TFA) as a yellow gum.
Step 3
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (4-piperidinylmethyl) piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (89.23 mg,137.13umol,8.25e-1 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (50 mg,166.26umol,1 eq) in DMSO (5 mL) was then added DIEA (107.44 mg,831.30umol,144.79ul,5 eq). The mixture was then stirred at 80℃for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:15% -35%,35 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (22.7 mg,26.94umol,16.20% yield, 95.05% purity) as a yellow solid.
Exemplary Synthesis of Compound 69
Step 1
To a mixture of tert-butyl 1-oxa-6-azaspiro [2.5] octane-6-carboxylate (910.28 mg,4.27mmol,1 eq.) in EtOH (10 mL) was added benzyl (3S) -3-methylpiperazine-1-carboxylate (1 g,4.27mmol,1 eq.) at 20deg.C under N2. The mixture was stirred at 90℃for 16 hours. TLC (dichloromethane: methanol=10:1, rf=0.57) and LCMS showed the reaction was complete. The residue was poured into water (10 mL). The aqueous phase was extracted with DCM (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (20 g,30ml/min,0% -100% (20 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ (1-tert-butoxycarbonyl-4-hydroxy-4-piperidinyl) methyl ] -3-methyl-piperazine-1-carboxylate (1.4 g,3.13mmol,73.29% yield) as a yellow oil.
Step 2
DAST (605.04 mg,3.75mmol,495.94uL,1.2 eq.) was added dropwise to a mixture of benzyl (3S) -4- [ (1-tert-butoxycarbonyl-4-hydroxy-4-piperidinyl) methyl ] -3-methyl-piperazine-1-carboxylate (1.4 g,3.13mmol,1 eq.) in DCM (10 mL) at-40 ℃. Then heated to 25 ℃ and stirred for 1h to give a colorless solution. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=1:1, rf=0.74) showed no starting material and a new spot. The reaction was cooled to 0 ℃ and quenched with aqueous NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with DCM (10 ml x 3). The combined organic layers were washed with brine (10 mL x 2) mL, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (12 g,0% -18% (5 min) ethyl acetate/petroleum ether, 18% (15 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) methyl ] -3-methyl-piperazine-1-carboxylate (764 mg,1.70mmol,54.35% yield) as a yellow oil.
Step 3
To a solution of benzyl (3S) -4- [ (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) methyl ] -3-methyl-piperazine-1-carboxylate (300 mg,667.32umol,1 eq.) in DCM (20 mL) was added TFA (228.26 mg,2.00mmol,148.22ul,3 eq.) at 20 ℃ in one portion. The mixture was stirred at 20℃for 30min to give a colorless solution. LCMS showed the reaction was complete. The residue was poured into NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with DCM (30 ml x 3). The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give benzyl (3S) -4- [ (4-fluoro-4-piperidinyl) methyl ] -3-methyl-piperazine-1-carboxylate (200 mg,475.04umol,71.19% yield, 83% purity) as a colorless oil.
Step 4
To a mixture of (3S) -4- [ (4-fluoro-4-piperidinyl) methyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (200 mg,572.34umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (211.41 mg,572.34umol,1 eq) in MeOH (10 mL) at 20 ℃ was added borane in one portion; 2-methylpyridine (122.44 mg,1.14mmol,2 eq.) and HOAc (1 mL). The mixture was stirred at 20℃for 1h to give a yellow solution. LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -20% MeOH/DCM) to give benzyl (3S) -4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-fluoro-4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylate (013 mg,530.72umol,92.73% yield) as a yellow solid.
Step 5
A mixture of (3S) -4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -4-fluoro-4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (370 mg,526.46umol,1 eq.) in TFA (7.70 g,67.53mmol,5mL,128.28 eq.) at 20℃in one portion. The mixture was stirred at 70 ℃ for 2 hours to give a yellow solution. LCMS showed the desired MS. The residue was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (529 mg,433.62umol,82.37% yield, 84% purity, 4 TFA) as a yellow gum.
Step 6
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (281.12 mg,274.32umol,1.1 eq, 4 TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (75 mg,249.38umol,1 eq) in DMSO (5 mL) at 20 ℃ was added DIEA single time (257.84 mg,2.00mmol,347.50ul,8 eq). The mixture was stirred at 80℃for 36h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow: 25mL/min; gradient time: 35min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (23.9 mg,28.46 mol,11.41% yield, 99.20% purity) as a yellow solid.
Exemplary Synthesis of Compound 70
Step 1
To a mixture of benzyl (3S) -3-methylpiperazine-1-carboxylate (500 mg,2.13mmol,1 eq.) and tert-butyl 4-formylpiperidine-1-carboxylate (546.17 mg,2.56mmol,1.2 eq.) in MeOH (10 mL) at 25℃was added NaOAc (175.07 mg,2.13mmol,1 eq.) in one portion; 2-methylpyridine (456.52 mg,4.27mmol,2 eq.) and HOAc (128.15 mg,2.13mmol,122.05uL,1 eq.). The mixture was stirred at 25℃for 1h. TLC (dichloromethane: methanol=10:1, rf=0.43, pma) showed that the reaction was complete. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10:1, rf= 0.43,0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -3-methyl-piperazine-1-carboxylate (69mg, 1.31mmol,61.43% yield, 82% purity) as a yellow oil.
Step 2
To a mixture of benzyl (3S) -4- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -3-methyl-piperazine-1-carboxylate (460 mg,1.60mmol,1 eq.) in DCM (10 mL) at 20℃was added TFA (3.08 g,27.01mmol,2mL,16.90 eq.) in one portion. The mixture was stirred at 20℃for 30min. LCMS showed the reaction was complete. The residue was concentrated in vacuo to give benzyl (3S) -3-methyl-4- (4-piperidinylmethyl) piperazine-1-carboxylate (1.3 g,1.52mmol,95.37% yield, 79% purity, 3 TFA) as a colorless oil.
Step 3
To a mixture of benzyl (3S) -3-methyl-4- (4-piperidinylmethyl) piperazine-1-carboxylate (1.3 g,1.93mmol,1 eq, 3 TFA) in EtOH (10 mL) was added tert-butyl 1-oxa-6-azaspiro [2.5] octane-6-carboxylate (823.30 mg,3.86mmol,2 eq) at 20deg.C under N2. The mixture was stirred at 90℃for 16 hours. TLC (dichloromethane: methanol=10:1, rf=0.57) and LCMS showed the reaction was complete. The residue was poured into water (10 mL). The aqueous phase was extracted with DCM (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (20 g,30ml/min,0% -100% (20 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ [1- [ (1-tert-butoxycarbonyl-4-hydroxy-4-piperidinyl) methyl ] -4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylate (370 mg,679.24umol,35.19% yield) as a yellow oil.
Step 4
DAST (130.91 mg,812.12umol,107.30uL,1.2 eq) was added dropwise to a mixture of benzyl (3S) -4- [ [1- [ (1-tert-butoxycarbonyl-4-hydroxy-4-piperidinyl) methyl ] -4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylate (368.65 mg,676.77umol,1 eq) in DCM (10 mL) at-40 ℃. Then heated to 25 ℃ and stirred for 1h to give a colorless solution. LCMS showed the desired MS. TLC (dichloromethane: methanol=20:1, rf=0.67) showed no starting material and a new spot. The reaction was cooled to 0 ℃ and quenched with aqueous NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with DCM (10 ml x 3). The combined organic layers were washed with brine (10 ml x 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (12 g,0% -18% (5 min) ethyl acetate/petroleum ether, 18% (15 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ [1- [ (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) methyl ] -4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylate (210 mg,384.11umol,56.76% yield) as a yellow oil.
Step 5
To a solution of benzyl (3S) -4- [ [1- [ (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) methyl ] -4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylate (210 mg,384.11umol,1 eq.) in DCM (5 mL) at 20 ℃ was added TFA (3.08 g,27.01mmol,2mL,70.33 eq.) in one portion. The mixture was stirred at 20℃for 30min to give a colorless solution. LCMS showed the reaction was complete. The residue was concentrated in vacuo to give benzyl (3S) -4- [ [1- [ (4-fluoro-4-piperidinyl) methyl ] -4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylate (310 mg,310.52umol,80.84% yield, 79% purity, 3 TFA) as a yellow oil.
Step 6
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (108.57 mg,393.07umol,1 eq.) and benzyl (3S) -4- [ [1- [ (4-fluoro-4-piperidinyl) methyl ] -4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylate (310 mg,393.07umol,1 eq., 3 TFA) in DMSO (5 mL) was added DIEA (508.00 mg,3.93mmol,684.64uL,10 eq.) at 20 ℃. The mixture was stirred at 100℃for 16h. LCMS showed the reaction was complete. The mixture was cooled to 20 ℃. The aqueous phase was extracted with ethyl acetate (3X 10 mL). The combined organic phases were washed with brine (2×10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -100% ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-fluoro-4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylate (150 mg,168.61umol,42.90% yield, 79% purity) as a yellow gum.
Step 7
A mixture of (3S) -4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-fluoro-4-piperidinyl ] methyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (150 mg,213.43umol,1 eq.) in TFA (3.12 g,27.38mmol,2.03mL,128.28 eq.) at 20℃in one portion. The mixture was stirred at 70 ℃ for 2 hours to give a yellow solution. LCMS showed the desired MS. The residue was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [ 4-fluoro-4- [ [4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione as a yellow gum (300 mg,194.94umol,91.34% yield, 74% purity, 5 TFA).
Step 8
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [ 4-fluoro-4- [ [4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (300 mg,527.54 mol,2.12 eq) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (75 mg,249.38 mol,1 eq) in DMSO (5 mL) was added DIEA (257.84 mg,2.00mmol,347.50ul,8 eq) at one time at 20 ℃. The mixture was stirred at 80℃for 24h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow: 25mL/min; gradient time: 35min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [ 4-fluoro-4- [ [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (25 mg,28.29umol,11.35% yield, 94.27% purity) as a yellow solid.
Exemplary Synthesis of Compound 71
Compound 71 was prepared in an analogous manner to compound 70 using 3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) -2H-indazole and 2- (2, 6-dioxopiperidin-3-yl) -5- (4- ((4- (((S) -2-methylpiperazin-1-yl) methyl) piperidin-1-yl) isoindoline-1, 3-dione.
Step 1
To a solution of 1-bromo-4, 5-difluoro-2-nitro-benzene (10 g,42.02mmol,1 eq.) and 1-methylcyclopropanol (3.03 g,42.02mmol,1 eq.) in DMF (50 mL) at 0deg.C was added NaH (2.52 g,63.03mmol,60% purity, 1.5 eq.) in one portion. Then heated to 20 ℃ and stirred for 2 hours to give a brown suspension. TLC showed the reaction was complete. The residue was poured into NH4Cl (100 mL) and stirred for 10min. The aqueous phase was extracted with ethyl acetate (3X 100 mL). The combined organic phases were washed with NH4Cl (2×100 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (column height: 80g,100-200 mesh silica gel, 100% (30 min) petroleum ether) to give 1-bromo-4-fluoro-5- (1-methylcyclopropoxy) -2-nitro-benzene (9 g,31.03mmol,73.84% yield) as a yellow oil.
Step 2
To a mixture of 1-bromo-4-fluoro-5- (1-methylcyclopropoxy) -2-nitro-benzene (9 g,31.03mmol,1 eq), K2CO3 (8.58 g,62.05mmol,2 eq) and Cs2CO3 (10.11 g,31.03mmol,1 eq) in 1, 4-dioxane (200 mL) was added trimethylcyclotriborane (19.47 g,77.56mmol,21.69mL,50% purity, 2.5 eq) and Pd (PPh 3) 4 (3.59 g,3.10mmol,0.1 eq) at 20 ℃ followed by heating to 100 ℃ and stirring for 16h to give a yellow solution. TLC showed the reaction was complete. The reaction was cooled to 20 ℃ and concentrated under vacuum. The residue was purified by silica gel chromatography (330 g,100-200 mesh silica gel, 100% (30 min) petroleum ether) to give 1-fluoro-4-methyl-2- (1-methylcyclopropoxy) -5-nitro-benzene (5.9 g,26.20mmol,84.44% yield) as a yellow oil.
Step 3
To a mixture of 1-fluoro-4-methyl-2- (1-methylcyclopropoxy) -5-nitrobenzene (5.9 g,26.20mmol,1 eq.) in EtOH (10 mL) at 20deg.C was added Pd/C (3 g,26.20mmol,10% purity, 1 eq.) and ammonium formate (19.82 g,314.37mmol,12 eq.) in one portion. The mixture was stirred at 20 ℃ for 2h to give a black solution. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=10:1, rf=0.23, uv 254 nm) showed no starting material and TLC showed new spots. The suspension was filtered through a pad of celite or silica gel and the pad or filter cake was washed with EtOH (100 ml x 3). The combined filtrates were concentrated. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=10:1, rf=0.16, 20g,0% -5% (3 min) ethyl acetate/petroleum ether, 5% (5 min) ethyl acetate/petroleum ether) to give 5-fluoro-2-methyl-4- (1-methylcyclopropoxy) aniline (5.3 g, crude) as a yellow oil.
Step 4
To a stirred solution of 5-fluoro-2-methyl-4- (1-methylcyclopropoxy) aniline (5.3 g,27.15mmol,1 eq.) in AcOH (60 mL) was added NaNO2 (2.06 g,29.86mmol,1.1 eq.) in H2O (15 mL) at 0 ℃. The reaction mixture was stirred at 20 ℃ for 16 hours. The reaction mixture changed color from yellow to brown. LCMS and TLC (petroleum ether: ethyl acetate=3:1, rf=0.16, uv 254 nm) showed the reaction was complete. To the residue was added saturated NaHCO3 solution (40 mL) and extracted with EA (40 mL). The combined organic layers were washed with brine (40 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (column height: 40g,100-200 mesh silica gel, 0% -30% (10 min) ethyl acetate/petroleum ether, 30% (20 min) ethyl acetate/petroleum ether) to give 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazole (1 g,4.85mmol,17.86% yield) as a yellow oil.
Step 5
To a mixture of 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazole (1 g,4.85mmol,1 eq.) in THF (40 mL) at 20deg.C was added N-cyclohexyl-N-methyl-cyclohexylamine (2.84 g,14.55mmol,3.09mL,3 eq.) and SEM-Cl (1.62 g,9.70mmol,1.72mL,2 eq.) in one portion. The mixture was stirred at 20 ℃ for 1h to give an orange suspension. TLC showed the reaction was complete. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (3X 30 mL). The combined organic phases were washed with brine (2×30 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -3% (20 min) ethyl acetate/petroleum ether, 3% -10% (10 min) ethyl acetate/petroleum ether) to give 2- [ [ 6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (1.1 g,3.27mmol,67.42% yield) as a yellow oil.
Step 6
To a mixture of 2- [ [ 6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (1.1 g,3.27mmol,1 eq.) in THF (5 mL) was added N-BuLi (2.5 m,1.57mL,1.2 eq.) dropwise at 70 ℃. The mixture was then stirred at-20 ℃ for 5min, and ZnCl2 solution (1 m,4.90ml,1.5 eq.) was added dropwise at-70 ℃. The mixture was stirred at-40℃for 10min. A mixture of 4, 6-dichloropyrimidine (487.04 mg,3.27mmol,1 eq.) and Pd (PPh 3) 4 (188.89 mg,163.46 mol,0.05 eq.) in THF (5 mL) was stirred at 20deg.C for 30min and added to the solution. The cold bath was removed and the mixture was stirred at 20 ℃ for 10h to give a yellow solution. TLC (petroleum ether: ethyl acetate=3:1, rf=0.83) showed the reaction was complete. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (3X 20 mL). The combined organic phases were washed with brine (2×20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -5% (30 min) ethyl acetate/petroleum ether, 5% (60 min) ethyl acetate/petroleum ether) to give 2- [ [3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (271mg, 603.57umol,18.46% yield) as a yellow oil.
Step 7
To a solution of 2- [ [3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (271mg, 603.57umol,1 eq.) in DCM (2 mL) was added TFA (4.01 g,35.13mmol,2.60mL,58.21 eq.). The mixture was then stirred at 20℃for 30min. LCMS showed the reaction was complete. The reaction mixture was poured into H2O (10 mL). The aqueous phase was adjusted to pH 9 with solid NaHCO 3. The mixture was extracted with ethyl acetate (15 ml x 3). The organic phase was washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. To the crude product in dioxane (2 mL) was added NH 3H 2O (1 mL) and stirred at 20 ℃ for 1H. LCMS showed the desired MS. The mixture was then extracted with ethyl acetate (15 ml x 3). The organic phase was washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (0% -10% ethyl acetate/petroleum ether) to give 3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) -2H-indazole (20 mg,57.73umol,9.56% yield, 92% purity) as a white solid.
Exemplary Synthesis of Compound 72
Compound 72 was prepared in a similar manner to compound 71 using 3- (6-chloropyrimidin-4-yl) -5- (cyclopropyloxy) -2H-indazole and 2- (2, 6-dioxopiperidin-3-yl) -5- (4- ((4- (((S) -2-methylpiperazin-1-yl) methyl) piperidin-1-yl) isoindoline-1, 3-dione.
Exemplary Synthesis of Compound 73
Step 1
To a solution of 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (80 mg,173.31umol,1 eq.) and tert-butyl 4-formylpiperidine-1-carboxylate (55.44 mg,259.96umol,1.5 eq.) in MeOH (5 mL) was added AcOH (1 mL); 2-methylpyridine (18.54 mg,173.31umol,1 eq). The mixture was stirred at 25℃for 16 hours. LCMS showed the desired MS. The reaction mixture was concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (dichloromethane: methanol=10:1, 0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 4- [ [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (120 mg,160.27umol,92.48% yield, 88% purity) as a colorless oil.
Step 2
To a solution of tert-butyl 4- [ [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (120 mg,182.13umol,1 eq) in DCM (5 mL) was added TFA (3.08 g,27.01mmol,2mL,148.31 eq). The mixture was stirred at 25℃for 1 hour. LCMS showed the desired MS. The mixture was concentrated in vacuo to give 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- [ [1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (150 mg, crude) as a brown oil.
Step 3
To 4, 5-difluorophthalic acid (450 mg,2.23mmol,1 eq.) and 3-aminopiperidine-2, 6-dione; to a solution of hydrochloride (549.69 mg,3.34mmol,1.5 eq.) in AcOH (5 mL) was added NaOAc (547.92 mg,6.68mmol,3 eq.). The mixture was stirred at 120℃for 15 hours. LCMS showed the desired MS. The mixture was diluted with water (50 mL). The mixture was filtered and the filter cake was washed with water (50 mL). The mixture was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5, 6-difluoro-isoindoline-1, 3-dione (300 mg,948.30umol,42.59% yield, 93% purity) as a black solid.
Step 4
To a mixture of 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- [ [1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (50 mg,89.48umol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5, 6-difluoro-isoindoline-1, 3-dione (39.49 mg,134.23umol,1.5 eq.) in MeCN (2 mL) was added DIEA (371.00 mg,2.87mmol,0.5mL,32.08 eq.). The mixture was stirred at 60℃for 16h. LCMS showed complete consumption of reaction 1 and found the desired MS. The reaction mixture was concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; conditions: [ water (0.225% FA) -ACN ]; start B:0 end B:35; flow rate: 25mL/min; gradient time: 35min;100% B hold time: 3 min) to afford the residue as a green solid, 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-6- [4- [ [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (7.8 mg,9.18umol,10.26% yield, 98% purity).
Exemplary Synthesis of Compound 74
Step 1
To a solution of tert-butyl 2, 5-dihydropyrrole-1-carboxylate (5 g,29.55mmol,1 eq.) in DCM (50 mL) under nitrogen was added rhodium (ii) acetate dimer (320 mg,724.00umol,2.45e-2 eq.). Ethyl 2-diazoacetate (3.37 g,29.55mmol,1 eq.) dissolved in DCM (40 mL) was then added dropwise slowly over 1 hour. After addition, the reaction mixture was stirred at 25 ℃ for 12h. TLC (petroleum ether: ethyl acetate=10:1) showed several new spots. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% ethyl acetate/petroleum ether) to give (1 s,5s,6 r) -3-azabicyclo [3.1.0] hexane-3, 6-dicarboxylic acid O3-tert-butyl ester O6-ethyl ester (650 mg,2.55mmol,8.62% yield) as a colorless oil and (1 s,5r,6 s) -3-azabicyclo [3.1.0] hexane-3, 6-dicarboxylic acid O3-tert-butyl ester O6-ethyl ester (460 mg,1.80mmol,6.10% yield) as a pale yellow oil.
Step 2
To a solution of (1 s,5r,6 s) -3-azabicyclo [3.1.0] hexane-3, 6-dicarboxylic acid O3-tert-butyl ester O6-ethyl ester (770 mg,3.02mmol,1 eq.) in EtOH (6 mL) was added dropwise NaOH (2M, 3.02mL,2 eq.) at 0deg.C. After the addition, the reaction solution was stirred at 25 ℃ for 12h. TLC (petroleum ether: ethyl acetate=10:1) showed that the starting material was consumed and a new spot formed. The reaction mixture was concentrated under reduced pressure. The residue was diluted with water (10 mL) and washed with ethyl acetate (10 mL x 2). The aqueous phase was acidified to pH 2 with 2M HCl and extracted with ethyl acetate (10 ml x 2). The organic layer was dried over sodium sulfate and concentrated under reduced pressure to give (1 s,5r,6 s) -3-tert-butoxycarbonyl-3-azabicyclo [3.1.0] hexane-6-carboxylic acid (600 mg, crude) as a brown solid. The crude product was used directly in the next step.
Step 3
A solution of (1 s,5r,6 s) -3-tert-butoxycarbonyl-3-azabicyclo [3.1.0] hexane-6-carboxylic acid (600 mg,2.64mmol,1 eq.) in THF (10 mL) was cooled to-10deg.C. Boranes; tetrahydrofuran (1 m,5.28ml,2 eq.) was slowly added to the flask while maintaining the temperature below 0 ℃. The solution was warmed to 25 ℃ and stirred for 12h. TLC (PE: etoac=1:1) showed that the starting material was consumed and a new spot formed. The solution was cooled to 0 ℃ and 15% sodium hydroxide solution (10 mL) was added dropwise over a period of 5 minutes to control gas evolution. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (3×20 mL). The organic layer was washed with brine (30 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 50% ethyl acetate/petroleum ether) to give (1 s,5r,6 s) -6- (hydroxymethyl) -3-azabicyclo [3.1.0] hexane-3-carboxylic acid tert-butyl ester (350 mg,1.64mmol,62.16% yield) as a colorless oil.
Step 4
To a solution of tert-butyl (1 s,5r,6 s) -6- (hydroxymethyl) -3-azabicyclo [3.1.0] hexane-3-carboxylate (50 mg,234.44umol,1 eq.) in DCM (2 mL) was added DMP (198.87 mg,468.88umol,2 eq.). After the addition, the reaction mixture was stirred at 25 ℃ for 1h. TLC (petroleum ether: ethyl acetate=1:1) showed the reaction was complete. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give (1 s,5r,6 s) -6-formyl-3-azabicyclo [3.1.0] hexane-3-carboxylic acid tert-butyl ester (49 mg, crude) as a white solid. The crude product was used directly in the next step.
Step 5
To a solution of (1 s,5r,6 s) -6-formyl-3-azabicyclo [3.1.0] hexane-3-carboxylic acid tert-butyl ester (49 mg,231.94umol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindoline-1, 3-dione (101.94 mg,231.94umol,1 eq.) in MeOH (2 mL) and HOAc (0.2 mL) was added borane; 2-methylpyridine (49.62 mg,463.89 mol,2 eq). After addition, the reaction mixture was stirred at 25 ℃ for 12h. LCMS showed the desired MS. TLC (dichloromethane: methanol=10:1) showed several new spots. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% methanol in dichloromethane) to give (1 r,5s,6 s) -6- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -3-azabicyclo [3.1.0] hexane-3-carboxylic acid tert-butyl ester (100 mg,157.54umol,67.92% yield) as a yellow solid.
Step 6
To a solution of (1 r,5s,6 s) -6- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -3-azabicyclo [3.1.0] hexane-3-carboxylic acid tert-butyl ester (100 mg,157.54umol,1 eq.) in DCM (2 mL) was added TFA (1.54 g,13.51mmol,1mL,85.73 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 1h. LCMS showed the starting material was consumed and the desired MS formed. The reaction mixture was concentrated under reduced pressure to give 5- [4- [ [4- [ [ (1 s,5r,6 s) -3-azabicyclo [3.1.0] hex-6-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (84 mg, crude) as a yellow gum. The crude product was used directly in the next step.
Step 7
To a solution of 5- [4- [ [4- [ [ (1 s,5r,6 s) -3-azabicyclo [3.1.0] hex-6-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (84 mg,157.11umol,1.43 eq) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (33 mg,109.73umol,1 eq) in DMSO (2 mL) was added DIEA (70.91 mg,548.64umol,95.56ul,5 eq). After the addition, the reaction solution was stirred at 90 ℃ for 12h. LCMS showed starting material was consumed and the desired MS was found. The residue was diluted with dichloromethane (20 mL) and washed with water (10 mL x 2). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: phenomenex Luna C18 75 x 30mm x 3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,35 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (1 r,5s,6 s) -3- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -3-azabicyclo [3.1.0] hex-6-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (31.1 mg,38.72umol,35.28% yield, 99.46% purity) as a yellow solid.
Exemplary Synthesis of Compound 75
Compound 75 was prepared in an analogous manner to compound 74 using 3- (tert-butyl) ester 6-ethyl (1 r,5s,6 r) -3-azabicyclo [3.1.0] hexane-3, 6-dicarboxylic acid.
Exemplary Synthesis of Compound 76
Step 1
To a solution of 5- (1-methylcyclopropoxy) -3- (6-piperazin-1-ylpyrimidin-4-yl) -1H-indazole (80 mg,228.30umol,1 eq.) in HOAc (0.4 mL) and MeOH (4 mL) was added 4-formylpiperidine-1-carboxylic acid tert-butyl ester (97.38 mg,456.60umol,2 eq.) and borane; 2-methylpyridine (48.84 mg,456.60 mol,2 eq). After the addition, the reaction solution was stirred at 25 ℃ for 12h. LCMS (EB 12-770-P1D) showed the desired MS. TLC (ethyl acetate: petroleum ether=2:1) showed several new spots. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (2×10 mL). The combined organic layers were washed with brine (20 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give 4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]Tert-butyl piperidine-1-carboxylate (75 mg,133.93umol,58.66% yield, 97.8% purity).
Step 2
To a solution of tert-butyl 4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (75 mg,136.94umol,1 eq.) in DCM (3 mL) was added TFA (2.31 g,20.26mmol,1.5mL,147.94 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 1h. TLC (pure ethyl acetate) showed that the starting material was consumed and a new spot formed. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in DMC (1 mL) and treated with DIEA (0.5 mL). The mixture was concentrated under reduced pressure to give 5- (1-methylcyclopropoxy) -3- [6- [4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -1H-indazole (65 mg, crude) as a yellow gum. The crude product was used directly in the next step.
Step 3
To a solution of 5- (1-methylcyclopropoxy) -3- [6- [4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -1H-indazole (65 mg,145.23umol,1 eq) in MeOH (5 mL) and HOAc (0.5 mL) was added 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (55 mg,148.90umol,1.03 eq) and borane; 2-methylpyridine (40 mg,373.97umol,2.58 eq). After addition, the reaction mixture was stirred at 25 ℃ for 12h. LCMS (EB 12-774-P1B 1) showed consumption of starting material and formation of the desired MS. The reaction was diluted with water (10 mL) and extracted with ethyl acetate (8 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -35%;40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (37.7 mg,46.93umol,32.31% yield, 99.70% purity) as a yellow solid.
Exemplary Synthesis of Compound 77
Compound 77 was prepared in a similar manner to compound 68.
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (4-piperidinylmethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (199.64 mg,307.28 mol,1.18 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) -2H-indazole (83.00 mg,260.41 mol,1 eq) in DMSO (5 mL) at 20 ℃ was added DIEA (269.24 mg,2.08mmol,362.86ul,8 eq) in one portion. The mixture was stirred at 80℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow: 25mL/min; gradient time: 40min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (19 mg,22.91 mol,8.80% yield, 98.61% purity) as a yellow solid.
Exemplary Synthesis of Compound 78
Step 1
To a mixture of 4-piperidinylmethanol (2 g,17.37mmol,1 eq), 4-bromo-2-fluoro-pyridine (3.06 g,17.37mmol,1 eq) in DMSO (10 mL) was added K2CO3 (4.80 g,34.73mmol,2 eq), and the mixture was stirred at 100 ℃ under an atmosphere of N2 for 4h. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=3:1) showed a new main point. The reaction mixture was concentrated under reduced pressure to give a residue. The crude material was purified by flash chromatography on silica gel (0% -50% ethyl acetate/petroleum ether) to give [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methanol (4.3 g,15.70mmol,90.41% yield, 99% purity) as a white solid.
Step 2
To a mixture of [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methanol (2 g,7.38mmol,1 eq.) and TosCl (2.81 g,14.75mmol,2 eq.) in DCM (10 mL) was added TEA (1.49 g,14.75mmol,2.05mL,2 eq.) and DMAP (450.56 mg,3.69mmol,0.5 eq.) at 0 ℃ in one portion under N2. The mixture was stirred at 20℃for 1 hour. LCMS showed the desired MS. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 50% ethyl acetate/petroleum ether) to give 4-methylbenzenesulfonic acid [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methyl ester (3.12 g,6.38mmol,86.52% yield, 87% purity) as a white solid.
Step 3
To a mixture of 4-methylbenzenesulfonic acid [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methyl ester (3.12 g,7.34mmol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester (2.05 g,11.00mmol,1.5 eq.) in MeCN (20 mL) at 0 ℃ was added KI (6.09 g,36.68mmol,5 eq.) and DIEA (4.74 g,36.68mmol,6.39mL,5 eq.) in one portion. The mixture was stirred at 80℃for 12 hours. LCMS showed the desired MS. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give tert-butyl 4- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (480 mg,1.44mmol,19.66% yield, 72% purity) as a white solid.
Step 4
To a solution of tert-butyl 4- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (500 mg,1.14mmol,1 eq.) and 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl) -1,3, 2-dioxapentaborane (288.97 mg,1.14mmol,1 eq.) in dioxane (10 mL) was added KOAc (335.03 mg,3.41mmol,3 eq.) and Pd (dppf) Cl2 (83.26 mg,113.79 mol,0.1 eq.). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 12h. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 4- [ [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl) -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (1 g, crude) as a black oil.
Step 5
To a solution of tert-butyl 4- [ [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (553 mg,1.14mmol,1 eq.) in dioxane (10 mL) and H2O (2 mL) was added 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (350 mg,787.61umol,6.93e-1 eq.), na2CO3 (361.46 mg,3.41mmol,3 eq.) and Pd (dppf) Cl2 (83.18 mg,113.68umol,0.1 eq.). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 16h. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure to give tert-butyl 4- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (850 mg, crude) as a pale yellow gum.
Step 6
To a solution of tert-butyl 4- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (850 mg,1.26mmol,1 eq.) in DCM (2 mL) was added TFA (3.08 g,27.01mmol,2mL,21.51 eq.). After the addition, the reaction mixture was stirred at 25 ℃ for 2h. LCMS showed the desired MS. The reaction mixture was poured into H2O (10 mL). The aqueous phase was adjusted to ph=9 with solid NaHCO 3. The mixture was extracted with ethyl acetate (15 ml x 3). The organic phase was washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. To the crude product in dioxane (2 mL) was added NH 3H 2O (1 mL) and stirred at 20 ℃ for 1H. The mixture was then extracted with ethyl acetate (15 ml x 3). The organic phase was washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give 5- (1-methylcyclopropoxy) -3- [2- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] -4-pyridinyl ] -1H-indazole (720 mg, crude, TFA) as a yellow gum.
Step 7
To a solution of 5- (1-methylcyclopropoxy) -3- [2- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] -4-pyridinyl ] -1H-indazole (280 mg,626.98umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (150 mg,406.10umol,6.48e-1 eq) in MeOH (5 mL) were added AcOH (0.5 mL) and borane; 2-methylpyridine (67.06 mg, 626.98. Mu. Mol,1 eq). After addition, the reaction mixture was stirred at 25 ℃ for 16h. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (125.5 mg,146.86umol,23.42% yield, 99% purity, FA) as a yellow solid.
Exemplary Synthesis of Compound 79
Compound 79 was prepared in a similar manner to compound 69.
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- (piperazin-1-ylmethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (110.66 mg,199.51umol,1 eq), 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (60 mg,199.51umol,1 eq) in DMSO (5 mL) was added DIEA (25.78 mg,199.51umol,34.75ul,1 eq), and the mixture was stirred at 80 ℃ for 16 hours. LCMS showed complete consumption of starting material and found the desired MS. The reaction mixture was filtered and concentrated under reduced pressure. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (23.3 mg,28.17umol,14.12% yield, 99% purity) as a yellow solid.
Exemplary Synthesis of Compound 80
Step 1
To a mixture of 4-bromo-2-fluoro-pyridine (175.17 mg,995.36umol,1 eq) and 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carboxylic acid tert-butyl ester (300 mg,995.36umol,1 eq) in DMSO (10 mL) at 100 ℃ was added K2CO3 (687.84 mg,4.98mmol,70.14mL,5 eq) in one portion. The mixture was stirred at 100℃for 2h to give a yellow solution. LCMS showed the reaction was complete. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (50 mL). The aqueous phase was extracted with ethyl acetate (50 ml x 4). The combined organic phases were washed with brine (50 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=1:1, rf=0.56, 12g,0% -20% (10 min) ethyl acetate/petroleum ether, 20% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 4- [ [4- (4-bromo-2-pyridinyl) piperazin-1-yl ] methyl ] -4-fluoro-piperidine-1-carboxylate (234 mg,511.61umol,51.40% yield) as a yellow oil.
Step 2
To a mixture of tert-butyl 4- [ [4- (4-bromo-2-pyridinyl) piperazin-1-yl ] methyl ] -4-fluoro-piperidine-1-carboxylate (150 mg,327.96umol,1 eq), pin2B2 (166.56 mg,655.91umol,2 eq) and KOAc (96.56 mg,983.87umol,3 eq) in dioxane (10 mL) was added Pd (dppf) Cl2 (12.00 mg,16.40umol,0.05 eq) at 25 ℃ in one portion. The mixture was stirred at 100℃for 1h. TLC showed the reaction was complete. The mixture was cooled to 25 ℃, filtered and concentrated in vacuo to give [2- [4- [ (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -4-pyridinyl ] boronic acid (500 mg,284.16umol,86.65% yield, 24% purity) as a black oil.
Step 3
To a mixture of [2- [4- [ (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -4-pyridinyl ] boronic acid (500 mg,284.16 mol,24% purity, 1 eq), 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (138.90 mg,312.57 mol,1.1 eq) and Na2CO3 (90.35 mg,852.47 mol,3 eq) in 1, 4-dioxane (10 mL) and H2O (2 mL) at 25 ℃ was added Pd (dppf) Cl2 (31.19 mg,42.62 mol,0.15 eq) in one portion. The mixture was stirred at 100℃for 16 hours. LCMS showed the desired MS. The mixture was cooled to 25 ℃, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -20% ethyl acetate/petroleum ether) to give tert-butyl 4-fluoro-4- [ [4- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (210 mg, crude material) as a yellow gum.
Step 4
To a mixture of tert-butyl 4-fluoro-4- [ [4- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (205.85 mg,296.20umol,1 eq.) in MeOH (5 mL) was added HCl/EtOAc (4 m,222.15ul,3 eq.) in one portion at 20 ℃. The mixture was stirred at 65℃for 15min. LCMS showed the reaction was complete. The residue was poured into NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 3- [2- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -4-pyridinyl ] -5- (1-methylcyclopropoxy) -1H-indazole (137 mg,245.03umol,82.72% yield, 83.09% purity) as a yellow solid.
Step 5
To a mixture of 3- [2- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -4-pyridinyl ] -5- (1-methylcyclopropoxy) -1H-indazole (130 mg,279.82umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (103.36 mg,279.82umol,1 eq) in MeOH (5 mL) at 25 ℃ was added borane in one portion; 2-methylpyridine (59.86 mg, 559.65. Mu.mol, 2 eq.) and HOAc (16.80 mg, 279.82. Mu.mol, 16.00. Mu.L, 1 eq.). The mixture was stirred at 25℃for 16h. LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [ [4- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (60.9 mg,73.70umol,26.34% yield, 98.99% purity) as a yellow solid.
Exemplary Synthesis of Compound 81
Compound 81 was prepared in a similar manner to compound 80.
Exemplary Synthesis of Compound 82
Step 1
To a mixture of benzyl 4- [ (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) methyl ] piperazine-1-carboxylate (400 mg,918.42umol,1 eq) in DCM (3 mL) was added TFA (9.24 g,81.04mmol,6.00mL,88.24 eq) and the mixture was stirred at 25℃for 1 hour. TLC (petroleum ether: ethyl acetate=1:1) showed complete consumption of starting material and a new spot was found. The reaction mixture was concentrated under reduced pressure to give benzyl 4- [ (4-fluoro-4-piperidinyl) methyl ] piperazine-1-carboxylate (400 mg, crude, TFA) as a yellow oil.
Step 2
To a mixture of 1- (4-bromo-2-pyridinyl) piperidine-4-carbaldehyde (240.72 mg,894.41umol,1 eq), benzyl 4- [ (4-fluoro-4-piperidinyl) methyl ] piperazine-1-carboxylate (300 mg,894.41umol,1 eq) in MeOH (10 mL) was added AcOH (1 mL), borane; 2-methylpyridine (95.67 mg,894.41umol,1 eq.) the mixture was then stirred at 25℃for 1 hour. TLC (petroleum ether: ethyl acetate=1:1) showed complete consumption of starting material and several new spots were found. LCMS showed the desired MS. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% petroleum ether/ethyl acetate) to give benzyl 4- [ [1- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methyl ] -4-fluoro-4-piperidinyl ] methyl ] piperazine-1-carboxylate (480 mg,791.09umol,88.45% yield, 97% purity) as a colorless gum.
Step 3
To a solution of 4- [ [1- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methyl ] -4-fluoro-4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (480 mg,815.56umol,1 eq.) and 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl) -1,3, 2-dioxapentaborane (207.10 mg,815.56umol,1 eq.) in dioxane (10 mL) were added Pd (dppf) Cl2 (59.68 mg,81.56umol,0.1 eq.) and KOAc (240.12 mg,2.45mmol,3 eq.). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 12h. LCMS showed the desired MS. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give benzyl 4- [ [ 4-fluoro-1- [ [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (520 mg, crude) as a black solid.
Step 4
To a solution of 4- [ [ 4-fluoro-1- [ [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (518 mg,814.96 mol,1 eq), 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (362.15 mg,814.96 mol,1 eq) in dioxane (10 mL) and H2O (2 mL) was added Pd (dppf) Cl2 (59.63 mg,81.50 mol,0.1 eq) and Na2CO3 (86.38 mg,814.96 mol,1 eq). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 16h. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=1:1) showed several new spots. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% petroleum ether/ethyl acetate) to give benzyl 4- [ [ 4-fluoro-1- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (630 mg,579.57 mol,71.12% yield, 76% purity) as a brown oil.
Step 5
To a solution of 4- [ [ 4-fluoro-1- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (630 mg,762.60umol,1 eq.) in MeOH (2 mL) was added HCl/dioxane (4 m,2mL,10.49 eq.). The reaction mixture was stirred under N2 at 65 ℃ for 1h. The reaction mixture was filtered and concentrated under reduced pressure. After addition, the crude product was taken up in TFA (3.08 g,27.01mmol,2mL,35.42 eq.) and the reaction mixture was then stirred under N2 at 70℃for 1h. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure to give 3- [2- [4- [ [ 4-fluoro-4- (piperazin-1-ylmethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] -4-pyridinyl ] -5- (1-methylcyclopropoxy) -1H-indazole (428 mg, crude) as a brown oil.
Step 6
To a solution of 3- [2- [4- [ [ 4-fluoro-4- (piperazin-1-ylmethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] -4-pyridinyl ] -5- (1-methylcyclopropoxy) -1H-indazole (100 mg,178.02umol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (49.17 mg,178.02umol,1 eq.) in DMSO (3 mL) was added DIEA (2.23 g,17.22mmol,3mL,96.75 eq.). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 16h. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (10 mM NH4HCO 3) -ACN ]; B%:30% -80%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-1- [ [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (23 mg,28.12umol,15.80% yield, 100% purity) as a yellow solid.
Exemplary Synthesis of Compound 83
Step 1
To a solution of tert-butyl 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carboxylate (600 mg,1.99mmol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (604.86 mg,2.19mmol,1.1 eq.) in DMSO (10 mL) was added DIEA (771.84 mg,5.97mmol,1.04mL,3 eq.). The mixture was stirred at 120℃for 2 hours. TLC (petroleum ether: ethyl acetate=0:1, rf=0.3) showed no starting material and a new spot. The residue was diluted with H2O (50 mL) and extracted with ethyl acetate (40 mL. Times.3). The combined organic layers were washed with brine (3×30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% to 70% ethyl acetate/petroleum ether) to give 4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -4-fluoro-piperidine-1-carboxylic acid tert-butyl ester (1.0 g,1.70mmol,85.58% yield, 95% purity) as a yellow solid.
Step 2
To a solution of 4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -4-fluoro-piperidine-1-carboxylic acid tert-butyl ester (160 mg,286.94umol,1 eq.) in DCM (2 mL) was added TFA (3.08 g,27.01mmol,2mL,94.14 eq.). The mixture was stirred at 25℃for 1 hour. TLC (petroleum ether: ethyl acetate=0:1, rf=0.01) showed no starting material and a new spot. The residue was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (163 mg, crude, TFA) as a yellow gum.
Step 3
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (163 mg,285.20umol,1 eq, TFA) in DCM (5 mL) was added DIEA (184.30 mg,1.43mmol,248.38ul,5 eq) and stirred at 25 ℃ for 10min. The mixture was then concentrated. A solution of the residue tert-butyl 4-formylpiperidine-1-carboxylate (72.99 mg,342.25umol,1.2 eq.) in MeOH (10 mL) and HOAC (1 mL) was stirred at 25℃for 5min, followed by the addition of borane; 2-methylpyridine (61.01 mg,570.41umol,2 eq). The mixture was then stirred at 25 ℃ under N2 for 16.TLC (dichloromethane: methanol=10:1, rf=0.3) showed the reaction was complete. The reaction mixture was poured into H2O (20 mL). The mixture was extracted with ethyl acetate (30 ml x 3). The organic phase was washed with brine (20 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (0% -10% methanol in dichloromethane) to give 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -4-fluoro-1-piperidinyl ] methyl ] piperidine-1-carboxylic acid tert-butyl ester (180 mg,203.43umol,71.33% yield, 74% purity) as a yellow gum.
Step 4
To a solution of tert-butyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -4-fluoro-1-piperidinyl ] methyl ] piperidine-1-carboxylate (180 mg,274.91umol,1 eq.) in DCM (2 mL) was added TFA (3.08 g,27.01mmol,2mL,98.26 eq.). The mixture was stirred at 25℃for 2 hours. TLC (dichloromethane: methanol=10:1, rf=0.01) showed the reaction was complete. The residue was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (180 mg, crude, TFA) as a yellow gum.
Step 5
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (150 mg,224.32 mol,1 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -6- (1-methylcyclopropoxy) -1H-indazole (53.97 mg,179.46 mol,0.8 eq) in DMSO (5 mL) and DIEA (289.92 mg,2.24mmol,390.72ul,10 eq). The mixture was stirred at 80℃for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-1- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (38.8 mg,47.09umol,20.99% yield, 99.39% purity) as a yellow solid.
Exemplary Synthesis of Compound 84
Step 1
To a solution of benzyl 4- (1-tert-butoxycarbonylpiperidine-4-carbonyl) piperazine-1-carboxylate (2.3 g,5.33mmol,1 eq.) in DCM (12 mL) was added TFA (5.45 g,47.79mmol,3.54mL,8.97 eq.). The mixture was stirred at 25℃for 1 hour. LC-MS (EB 2049-70-P1A) showed complete consumption of reactant 1 and detection of a main peak with the desired m/z. The reaction mixture was concentrated under reduced pressure to remove the solvent, then the residue was quenched with saturated NaHCO3 (30 mL) and extracted with EA (30 mL). The organic layer was washed with water (30 ml×2), brine (30 ml×2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give benzyl 4- (piperidine-4-carbonyl) piperazine-1-carboxylate (1.9 g,4.27mmol,80.03% yield, TFA) as a yellow oil. The crude product was used in the next step without further purification.
Step 2
To a solution of benzyl 4- (piperidine-4-carbonyl) piperazine-1-carboxylate (220 mg,663.83 mol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (245.20 mg,663.83 mol,1 eq) in MeOH (5 mL) and AcOH (2 mL) and MeOH (5 mL) was added borane; 2-methylpyridine (142.01 mg,1.33mmol,2 eq.). The mixture was stirred at 25℃for 16 hours. LC-MS (EB 2049-71-P1B) showed complete consumption of reactant 1 and detection of a major peak with the desired m/z. The reaction mixture was concentrated under reduced pressure to remove the solvent. The residue was purified by flash chromatography on silica gel (0% -10% MeOH/DCM) to give benzyl 4- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperidine-4-carbonyl ] piperazine-1-carboxylate (202 mg,294.99umol,44.44% yield) as a yellow oil.
Step 3
To a solution of benzyl 4- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperidine-4-carbonyl ] piperazine-1-carboxylate (202 mg,294.99umol,1 eq.) in TFA (3 mL). The mixture was stirred at 70℃for 1 hour. TLC indicated complete consumption of reactant 1 and formation of a new spot. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (piperazine-1-carbonyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (400 mg,248.83umol,84.35% yield, 91% purity, 8 TFA) as a pale yellow oil. The crude product was used in the next step without further purification.
Step 4
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (piperazine-1-carbonyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (400 mg,273.44umol,1 eq., 8 TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (82.24 mg,273.44umol,1 eq.) in DMSO (3 mL) was added DIEA (353.40 mg,2.73mmol,476.29ul,10 eq.). The mixture was stirred at 80℃for 16 hours. LCMS showed complete consumption of reactant 1 and detection of one major peak with the desired m/z. The reaction mixture was concentrated under reduced pressure to remove the solvent. The crude product was purified by reverse phase HPLC (column: phenomena Luna C18 75X 30mm X3 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:5% -35%,35 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazine-1-carbonyl ] -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (17.6 mg,19.86umol,7.26% yield, 91.967% purity) as a white solid.
Exemplary Synthesis of Compound 85
Step 1
To a solution of 1-tert-butoxycarbonylpiperidine-4-carboxylic acid (10.41 g,45.40mmol,1 eq.) in DMF (100 mL) was added DIEA (17.60 g,136.20mmol,23.72mL,3 eq.) and HATU (22.44 g,59.02mmol,1.3 eq.). The mixture was stirred at 25℃for 0.5h. Benzyl piperazine-1-carboxylate (10 g,45.40mmol,8.77ml,1 eq.) was added to the reaction solution. The mixture was stirred at 25℃for 2h. TLC (petroleum ether/ethyl acetate=1/1, rf=0.17) indicated no residue of reactant 1 and a major new spot of lower polarity was detected. The reaction mixture was diluted with water (20 mL x 3) and extracted with EA (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by silica gel column chromatography (0% -60% (15 min) ethyl acetate/petroleum ether, 60% (10 min) ethyl acetate/petroleum ether) to give benzyl 4- (1-tert-butoxycarbonylpiperidine-4-carbonyl) piperazine-1-carboxylate (13 g,30.13mmol,66.36% yield) as a yellow oil.
Step 2
To a solution of benzyl 4- (1-tert-butoxycarbonylpiperidine-4-carbonyl) piperazine-1-carboxylate (3 g,6.95mmol,1 eq.) in EtOAc (50 mL) under N2 was added Pd/C (500 mg,6.95mmol,10% purity, 1 eq.). The suspension was degassed under vacuum and purged several times with H2. The mixture was stirred at 25℃under H2 (15 psi) for 3 hours. TLC (petroleum ether/ethyl acetate=1/1, rf=0) indicated no residue of reactant 1 and a major new spot of lower polarity was detected. The reaction mixture was filtered, and the filtrate was concentrated to give tert-butyl 4- (piperazine-1-carbonyl) piperidine-1-carboxylate (1.8 g,6.05mmol,87.06% yield) as a yellow oil.
Step 3
To a solution of tert-butyl 4- (piperazine-1-carbonyl) piperidine-1-carboxylate (900 mg,3.03mmol,1 eq) in DMSO (10 mL) was added K2CO3 (1.25 g,9.08mmol,3 eq) and 4-bromo-2-fluoro-pyridine (692.37 mg,3.93mmol,1.3 eq). The mixture was stirred at 100℃for 16 hours. LC-MS showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 53% of the desired compound was detected. The reaction mixture was diluted with water (20 mL) and extracted with EA (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by column chromatography on silica gel (column height: 20g,100-200 mesh silica gel, 0% -20% (15 min) ethyl acetate/petroleum ether, 20% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 4- [4- (4-bromo-2-pyridinyl) piperazine-1-carbonyl ] piperidine-1-carboxylate (1.1 g,2.43mmol,80.17% yield) as a yellow oil.
Step 4
4- [4- (4-bromo-2-pyridinyl) piperazine-1-carbonyl ] piperidine-1-carboxylic acid tert-butyl ester (300 mg,661.71 mol,1 eq.) 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl) -1,3, 2-dioxapentaborane (252.05 mg,992.56 mol,1.5 eq.) and A mixture of Pd (dppf) Cl2 (72.63 mg,99.26umol,0.15 eq), KOAc (194.82 mg,1.99mmol,3 eq) and 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxapentalan-2-yl) -1,3, 2-dioxapentalan (252.05 mg,992.56umol,1.5 eq) in dioxane (15 mL) was degassed and purged 3 times with N2, the mixture was then stirred at 90 ℃ under an atmosphere of N2 for 12 hours. LC-MS (EB 134-911-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 61% of the desired compound was detected. The reaction mixture was diluted with water (20 mL) and extracted with EA (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give [2- [4- (1-tert-butoxycarbonylpiperidine-4-carbonyl) piperazin-1-yl ] -4-pyridinyl ] boronic acid (260 mg,444.42umol,67.16% yield, 71.5% purity) as a black oil.
Step 5
A mixture of [2- [4- (1-tert-butoxycarbonylpiperidine-4-carbonyl) piperazin-1-yl ] -4-pyridinyl ] boronic acid (140 mg,334.69umol,1 eq), 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (148.73 mg,334.69umol,1 eq), pd (dppf) Cl2 (24.49 mg,33.47umol,0.1 eq), na2CO3 (106.42 mg,1.00mmol,3 eq) in dioxane (6 mL) and H2O (1 mL) was degassed and purged 3 times with N2, then the mixture was stirred under an atmosphere of N2 at 90℃for 3 hours. LC-MS (EB 134-917-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 47% of the desired compound was detected. The reaction mixture was diluted with water (20 mL) and extracted with EA (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by silica gel column chromatography (0% -60% (25 min) ethyl acetate/petroleum ether, 60% (10 min) ethyl acetate/petroleum ether) to give 4- [4- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] piperazine-1-carbonyl ] piperidine-1-carboxylic acid tert-butyl ester (130 mg,174.98umol,52.28% yield, 93% purity) as a yellow gum.
Step 6
To a solution of 4- [4- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] piperazine-1-carbonyl ] piperidine-1-carboxylic acid tert-butyl ester (130 mg,188.15umol,1 eq.) in MeOH (2 mL) was added HCl/dioxane (4 m,1.44mL,30.71 eq.). The mixture was stirred at 25℃for 2 hours. LC-MS (EB 134-919-P1B) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 80% of the desired compound was detected. The mixture was quenched with saturated NaHCO3 (30 mL) and extracted with EA (30 mL). The organic layer was washed with water (30 ml×2), brine (30 ml×2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give [4- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] piperazin-1-yl ] - (4-piperidinyl) methanone as a yellow gum (100 mg,187.81umol,99.82% yield, 86.5% purity).
Step 7
To a solution of [4- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] piperazin-1-yl ] - (4-piperidinyl) methanone (100 mg,217.12umol,1 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (88.22 mg,238.83umol,1.1 eq.) in MeOH (3 mL) and AcOH (0.3 mL) was added borane; 2-methylpyridine (116.12 mg,1.09mmol,5 eq.). The mixture was stirred at 25℃for 12 hours. LC-MS (EB 134-920-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 61% of the desired compound was detected. The resulting product was filtered to remove insoluble materials. The impure product was purified by preparative HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -35%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [4- [4- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] -2-pyridinyl ] piperazine-1-carbonyl ] -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (101 mg,122.97umol,56.64% yield, 99.101% purity) as a yellow solid.
Exemplary Synthesis of Compound 86
Compound 86 was prepared in a similar manner to compound 84.
Exemplary Synthesis of Compound 87
Step 1
To a mixture of 1-tert-butoxycarbonylpiperidine-4-carboxylic acid (3.80 g,16.57mmol,1 eq), HATU (9.45 g,24.86mmol,1.5 eq), DIEA (6.43 g,49.71mmol,8.66mL,3 eq) in DCM (10 mL) was added benzyl 4- (4-piperidinylmethyl) piperazine-1-carboxylate (5.26 g,16.57mmol,1 eq) and the mixture was stirred at 25 ℃ for 1 hour. LCMS showed the desired MS. TLC (dichloromethane: methanol=10:1) showed complete consumption of starting material and several new spots were found. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 30% dichloromethane/methanol) to give benzyl 4- [ [1- (1-tert-butoxycarbonylpiperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (11 g,15.19mmol,91.66% yield, 73% purity) as a yellow gum.
Step 2
To a mixture of benzyl 4- [ [1- (1-tert-butoxycarbonylpiperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (3.6 g,6.81mmol,1 eq.) in DCM (3 mL) was added TFA (33.26 g,291.73mmol,21.60mL,42.84 eq.) and the mixture was stirred at 25℃for 1 hour. TLC (dichloromethane: methanol=10:1) showed a new spot. The reaction mixture was concentrated under reduced pressure to give benzyl 4- [ [1- (piperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (3 g, crude, TFA) as a yellow oil.
Step 3
To a mixture of benzyl 4- [ [1- (piperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (1.4 g,3.27mmol,1 eq.) and 4-bromo-2-fluoro-pyridine (574.90 mg,3.27mmol,1 eq.) in DMSO (5 mL) was added DIEA (1.27 g,9.80mmol,1.71mL,3 eq.) and the mixture was stirred at 80 ℃ for 16 hours. LCMS showed complete consumption of starting material and found the desired MS. TLC (dichloromethane: methanol=10:1) showed several new spots. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (0% to 30% dichloromethane/methanol) to give benzyl 4- [ [1- [1- (4-bromo-2-pyridinyl) piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (1.22 g,1.11mmol,33.86% yield, 53% purity) as a yellow gum.
Step 4
To a solution of benzyl 4- [ [1- [1- (4-bromo-2-pyridinyl) piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (300 mg,513.22umol,1 eq.) and 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl) -1,3, 2-dioxapentaborane (130.33 mg,513.22umol,1 eq.) in dioxane (10 mL) were added Pd (dppf) Cl2 (37.55 mg,51.32umol,0.1 eq.) and KOAc (151.10 mg,1.54mmol,3 eq.). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 12h. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure to give benzyl 4- [ [1- [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (400 mg, crude) as a black solid.
Step 5
To a solution of 4- [ [1- [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (142.13 mg,225.03umol,1 eq), 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (100 mg,225.03umol,1 eq) in dioxane (10 mL) and H2O (2 mL) was added Pd (dppf) Cl2 (16.47 mg,22.50umol,0.1 eq) and Na2CO3 (71.55 mg,675.09umol,3 eq). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 16h. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=1:1) showed several new spots. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 30% methanol/dichloromethane) to give benzyl 4- [ [1- [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (90 mg,53.64umol,23.84% yield, 49% purity) as a black solid.
Step 6
To a solution of benzyl 4- [ [1- [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (90 mg,109.47 mol,1 eq.) in TFA (12.48 mg,109.47 mol,8.11ul,1 eq.). After addition, the reaction mixture was stirred at 70 ℃ under N2 for 0.5h. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure to give [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] - [4- (piperazin-1-ylmethyl) -1-piperidinyl ] methanone (80 mg, crude TFA) as a brown oil.
Step 7
To a mixture of [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] - [4- (piperazin-1-ylmethyl) -1-piperidinyl ] methanone (50 mg,89.65umol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (24.76 mg,89.65umol,1 eq.) in DMSO (3 mL) was added DIEA (11.59 mg,89.65umol,15.62ul,1 eq.) and the mixture was stirred at 100 ℃ for 16 hours. LCMS showed complete consumption of starting material and found the desired MS. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (10 mM NH4HCO 3) -ACN;% B: 20% -70%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (13.5 mg,16.41umol,18.31% yield, 98.95% purity) as a yellow solid.
Exemplary Synthesis of Compound 88
Step 1
To a solution of 1-benzyloxycarbonyl piperidine-4-carboxylic acid (5.59 g,21.24mmol,1.2 eq.) in DCM (60 mL) was added HATU (8.75 g,23.02mmol,1.3 eq.) and the mixture was stirred under N2 at 25℃for 1 h. And then tert-butyl 4- (4-piperidylmethyl) piperidine-1-carboxylate (5 g,17.70mmol,1 eq.) and DIEA (4.58 g,35.41mmol,6.17ml,2 eq.) were added to the above mixture. The mixture was stirred at 25 ℃ under N2 for 15 hours. TLC (petroleum ether: ethyl acetate=1:2, rf=0.46, pma) showed that a new main spot formed. To the reaction mixture was added 100mL of water and extracted with DCM (100 mL x 3). The combined organic layers were washed with brine (100 ml x 3), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel (0% -55% ethyl acetate/petroleum ether). The compound 4- [ [1- (1-benzyloxycarbonyl piperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperidine-1-carboxylic acid tert-butyl ester (7.6 g, crude) was obtained as a pale yellow gum.
Step 2
To a solution of tert-butyl 4- [ [1- (1-benzyloxycarbonyl piperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperidine-1-carboxylate (2.1 g,3.98mmol,1 eq.) in DCM (20 mL) was added HCl/dioxane (4 m,10mL,10.05 eq.) and the mixture stirred at 25 ℃ for 2 hours. TLC (petroleum ether: ethyl acetate=1:1, pma, plate 1) showed the formation of new spots. The reaction mixture was concentrated under reduced pressure to give a residue. The compound 4- [4- (4-piperidinylmethyl) piperidine-1-carbonyl ] piperidine-1-carboxylic acid benzyl ester (1.45 g, crude, HCl) was obtained as a white solid.
Step 3
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (200 mg,724.06umol,1 eq) and 4- [4- (4-piperidinylmethyl) piperidine-1-carbonyl ] piperidine-1-carboxylic acid benzyl ester (503.99 mg,1.09mmol,1.50 eq, HCl) in DMSO (5 mL) was added DIEA (467.90 mg,3.62mmol,630.59ul,5 eq) and the mixture was stirred at 90 ℃ for 16 hours. TLC (petroleum ether: ethyl acetate=3:1, rf=0.49, uv=254 nm, plate 1) showed that new spots formed. To the reaction mixture was added 30mL of water and extracted with EtOAc (30 mL x 3). The combined organic layers were washed with brine (30 ml x 3), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel (0% -10% ethyl acetate/petroleum ether). The compound 4- [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperidine-1-carbonyl ] piperidine-1-carboxylic acid benzyl ester (210 mg,270.26umol,37.33% yield, 88% purity) was obtained as a yellow solid.
Step 4
To a solution of benzyl 4- [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperidine-1-carbonyl ] piperidine-1-carboxylate (210 mg,307.11umol,1 eq.) in TFA (4.62 g,40.52mmol,3ml,131.93 eq.) the mixture was then stirred at 80 ℃ for 1 hour. LCMS showed about 88.53% compound and starting material was completely consumed. The reaction mixture was concentrated under reduced pressure to give a residue. The compound 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- (piperidine-4-carbonyl) -4-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (200 mg,155.11umol,50.51% yield, 78% purity, 4 TFA) was obtained as a pale yellow gum.
Step 5
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- (piperidine-4-carbonyl) -4-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (200 mg,198.86umol,1 eq., 4 TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (59.80 mg,198.86umol,1 eq.) in DMSO (3 mL) was added DIEA (257.01 mg,1.99mmol,346.37ul,10 eq.) and the mixture was stirred at 90 ℃ for 16 hours. LCMS showed about 50.5% of the desired compound and the starting material was completely consumed. The residue was purified by preparative HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:10% -60%,40 min). The compound 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (45 mg,53.27umol,26.79% yield, 96.346% purity) was obtained as a yellow solid.
Exemplary Synthesis of Compound 89
Compound 89 was prepared in a similar manner to compound 82 to give tert-butyl 4- [ [1- [1- (4-bromo-2-pyridinyl) piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperidine-1-carboxylate.
Step 1
To a solution of tert-butyl 4- [ [1- (1-benzyloxycarbonyl piperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperidine-1-carboxylate (2 g,3.79mmol,1 eq.) in EtOH (20 mL) and EtOAc (20 mL) under N2 was added Pd/C (0.54 g,10% purity). The suspension was degassed under vacuum and purged several times with H2 (15 psi). The reaction mixture was stirred at 25℃for 1h. TLC (petroleum ether: ethyl acetate=1:2) showed the reaction was complete. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 4- [ [1- (piperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperidine-1-carboxylate (1.45 g,3.44mmol,90.89% yield, 93.5% purity) as a white gum.
Step 2
To a solution of tert-butyl 4- [ [1- (piperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperidine-1-carboxylate (500 mg,1.27mmol,1 eq.) and 4-bromo-2-fluoro-pyridine (290.66 mg,1.65mmol,1.3 eq.) in DMSO (8 mL) was added K2CO3 (526.75 mg,3.81mmol,3 eq.). After the addition, the reaction solution was stirred at 100 ℃ for 12h. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=1:1) showed several spots. After cooling, the reaction mixture was filtered, and the filtrate was diluted with ethyl acetate (20 mL) and washed with brine (20 mL x 3). The organic layer was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 65% ethyl acetate/petroleum ether) to give tert-butyl 4- [ [1- [1- (4-bromo-2-pyridinyl) piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperidine-1-carboxylate (300 mg,510.97umol,40.22% yield, 93.6% purity) as a white solid.
Exemplary Synthesis of Compound 90
Compound 90 was prepared in a similar manner to compound 84.
Exemplary Synthesis of Compound 91
Compound 91 was prepared in a similar manner to compound 82.
Exemplary Synthesis of Compound 92
Step 1
To a solution of tert-butyl 4- [ [1- (piperidine-4-carbonyl) -4-piperidinyl ] methyl ] piperidine-1-carboxylate (300 mg,762.27umol,1 eq) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (210.55 mg,762.27umol,1 eq) in DMSO (5 mL) was added DIEA (492.59 mg,3.81mmol,663.87ul,5 eq) after the addition the mixture was stirred at 100 ℃ for 16 hours LCMS showed the desired MS. the residue was poured into water (20 mL), the aqueous phase was extracted with ethyl acetate (20 mL x 3), the combined organic phases were washed with brine (20 mL x 2), dried with anhydrous Na2SO4, filtered and concentrated in vacuo, the residue was purified by silica gel chromatography (4 g,0% -80% (12 min) ethyl acetate/petroleum ether, 80% (10 min) ethyl acetate/petroleum ether to give [ 4- [1- [ 6-dioxo-3-piperidinyl ] piperidine-1-carboxylate as a yellow solid ([ 4.81 mg, 663.87ul,5 eq) with the desired yield of MS.).
Step 2
To a solution of tert-butyl 4- [ [1- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbonyl ] -4-piperidinyl ] methyl ] piperidine-1-carboxylate (200 mg,307.80umol,1 eq.) in DCM (3 mL) was added TFA (1.23 g,10.80mmol,0.8mL,35.10 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 1h. TLC showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- (4-piperidinylmethyl) piperidine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (150 mg,198.89umol,64.62% yield, 88% purity, TFA) as a brown solid.
Step 3
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- (4-piperidinylmethyl) piperidine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (118.80 mg,216.13umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (65 mg,216.13umol,1 eq.) in DMSO (3 mL) was added DIEA (371.00 mg,2.87mmol,0.5mL,13.28 eq.). After addition, the mixture was stirred at 90 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 10% -50%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperidine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (13.1 mg,15.97umol,7.39% yield, 99.2% purity) as a yellow solid.
Exemplary Synthesis of Compound 93
Compound 93 was prepared in a similar manner to compound 82.
Exemplary Synthesis of Compound 94
Compound 94 was prepared in a similar manner to compound 92.
Exemplary Synthesis of Compound 95
Compound 95 was prepared in a similar manner to compound 82.
Exemplary Synthesis of Compound 96
Step 1
To a mixture of 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (257.40 mg,597.21 mol,1 eq) and tert-butyl 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carboxylate (180 mg,597.21 mol,1 eq) in DMSO (5 mL) was added Et3N (181.29 mg,1.79mmol,249.37ul,3 eq) at once at 25 ℃. The mixture was stirred at 100℃for 1h to give a yellow solution. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃ and concentrated under reduced pressure at 25 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=3:1, rf= 0.43,0% -100% (15 min) ethyl acetate/petroleum ether) to give tert-butyl 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (300 mg,431.07umol,72.18% yield) as a yellow solid.
Step 2
To a mixture of tert-butyl 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (300 mg,431.07umol,1 eq.) in MeOH (5 mL) was added HCl/EtOAc (4 m,2mL,18.56 eq.) in one portion followed by stirring at 65 ℃ for 30min. TLC showed complete consumption of starting material. The mixture was concentrated in vacuo to give 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (199mg, 418.89umol,97.17% yield, 98% purity) as a yellow solid.
Step 3
HATU (73.50 mg,193.31umol,1 eq) was added in one portion to a mixture of 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carboxylic acid (74.50 mg,193.31umol,1 eq) and 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1H-indazole (90 mg,193.31umol,1 eq) in DMF (5 mL) at 0 ℃. The mixture was stirred at 0℃for 3 hours. LCMS showed the desired product MS. The resulting product was poured into H2O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by preparative HPLC (column: phenomenex Luna C, 18, 75X 30mm X3 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:15% -45%,35 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] methyl ] piperidin-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (68.8 mg,82.49umol,42.67% yield, 99.87% purity) as a yellow solid.
Exemplary Synthesis of Compound 97
Compound 97 was prepared in a similar manner to compound 87.
Step 1
To a mixture of [2- [4- [ (1-tert-butoxycarbonyl-4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -4-pyridinyl ] boronic acid (280 mg,225.43umol,34% purity, 1 eq), 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (110.20 mg,247.98umol,1.1 eq) and Na2CO3 (71.68 mg,676.30umol,3 eq) in 1, 4-dioxane (10 mL) and H2O (2 mL) was added at one time Pd (dppf) Cl2 (24.74 mg,33.81umol,0.15 eq) at 25 ℃. The mixture was stirred at 100℃for 16 hours. LCMS showed the desired MS. The mixture was cooled to 25 ℃, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -20% ethyl acetate/petroleum ether) to give tert-butyl 4-fluoro-4- [ [4- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (134 mg,176.04umol,78.09% yield, 91.3% purity) as a yellow gum.
Step 2
To a mixture of tert-butyl 4-fluoro-4- [ [4- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (134 mg,192.82umol,1 eq.) in MeOH (5 mL) was added HCl/EtOAc (4 m,48.20ul,1 eq.) in one portion followed by stirring at 65 ℃ for 30min to give a yellow solution. TLC showed complete consumption of starting material. The mixture was concentrated in vacuo to give 3- [2- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -4-pyridinyl ] -5- (1-methylcyclopropoxy) -2H-indazole (76.3 mg,146.17umol,75.81% yield, 89% purity) as a yellow oil.
Step 3
HATU (60.48 mg,159.07umol,1 eq) was added in one portion to a mixture of 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carboxylic acid (61.30 mg,159.07umol,1 eq) and 3- [2- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -4-pyridinyl ] -5- (1-methylcyclopropoxy) -1H-indazole (73.9 mg,159.07umol,1 eq) in DMF (5 mL) at 0 ℃. The mixture was stirred at 0 ℃ and stirred for 3 hours. LCMS showed the desired MS. The resulting product was poured into H2O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by preparative HPLC (column: phenomenex Luna C, 75X 30mm X3 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:20% -50%,35 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ 4-fluoro-4- [ [4- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] piperazin-1-yl ] methyl ] piperidine-1-carbonyl ] -1-piperidinyl ] isoindoline-1, 3-dione (9.3 mg,11.13umol,7.00% yield, 99.59% purity) as a yellow solid.
Exemplary Synthesis of Compound 98
Step 1
Cs2CO3 (7.41 g,22.73mmol,2 eq.) was added in one portion to a mixture of 4- (dimethoxymethyl) piperidine (1.81 g,11.36mmol,1 eq.) and 2-bromo-4-fluoro-pyridine (2 g,11.36mmol,1 eq.) in MeCN (50 mL) at 25 ℃ under N2. The mixture was stirred at 50℃for 3 hours. The mixture was filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 10/1) to give 2-bromo-4- [4- (dimethoxymethyl) -1-piperidinyl ] pyridine (3.1 g,9.24mmol,81.35% yield, 94% purity) as a yellow solid.
Step 2
To a mixture of 2-bromo-4- [4- (dimethoxymethyl) -1-piperidinyl ] pyridine (1.5 g,4.76mmol,1 eq.) in THF (10 mL) was added HCl (2 m,10mL,4.20 eq.) in one portion at 25 ℃. The mixture was stirred at 25℃for 2 hours. TLC showed the reaction was complete. The mixture was poured into saturated aqueous NaHCO3 (30 mL). The aqueous phase was extracted with ethyl acetate (30 ml x 3). The combined organic phases were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 1- (2-bromo-4-pyridinyl) piperidine-4-carbaldehyde (1.3 g, crude) as a yellow oil.
Step 3
To a mixture of 1- (2-bromo-4-pyridinyl) piperidine-4-carbaldehyde (1.3 g,4.83mmol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester (989.60 mg,5.31mmol,1.1 eq.) in MeOH (20 mL) and HOAc (2 mL) at 25 ℃ was added borane in one portion; 2-methylpyridine (1.03 g,9.66mmol,2 eq.). The mixture was stirred at 25℃for 12 hours. The mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 1/1) to give tert-butyl 4- [ [1- (2-bromo-4-pyridinyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (2.0 g,4.55mmol,94.23% yield) as a white solid.
Step 4
Pd (PPh 3) 2Cl2 (11.85 mg,16.88umol,0.1 eq) was added in one portion to a mixture of trimethyl (trimethylstannyl) stannane (110.59 mg,337.55umol,69.99uL,2 eq.) and 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (75.00 mg,168.77umol,1 eq.) in toluene (3 mL) at 25 ℃. The mixture was stirred at 140℃for 1 hour. TLC showed the reaction was complete. The mixture was filtered and concentrated in vacuo to give trimethyl- [2- [ [5- (1-methylcyclopropoxy) -3-trimethylstannanyl-indazol-1-yl ] methoxy ] ethyl ] silane (80 mg,101.39umol,60.08% yield, 61% purity) as a yellow solid.
Step 5
To a mixture of trimethyl- [2- [ [5- (1-methylcyclopropoxy) -3-trimethylstannyl-indazol-1-yl ] methoxy ] ethyl ] silane (80.00 mg,166.22 mol,1 eq) and tert-butyl 4- [ [1- (2-bromo-4-pyridinyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (146.07 mg,332.44 mol,2 eq) in THF (5 mL) at 25 ℃ was added LiCl (35.23 mg,831.10 mol,17.02ul,5 eq), cuI (4.75 mg,24.93 mol,0.15 eq) and Pd (PPh 3) 4 (28.81 mg,24.93 mol,0.15 eq) in one portion. The mixture was stirred by MW at 100deg.C for 2 hours. The mixture was filtered and concentrated in vacuo. The residue was purified by preparative TLC (petroleum ether/ethyl acetate=1/1) to give 4- [ [1- [2- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -4-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylic acid tert-butyl ester (200 mg, crude material) as a yellow solid.
Step 6
To a mixture of tert-butyl 4- [ [1- [2- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -4-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (200 mg,295.44umol,1 eq.) in DCM (5 mL) was added HCl/dioxane (4 m,5mL,67.70 eq.) in portions at 25 ℃. The mixture was stirred at 25℃for 2 hours. TLC showed the reaction was complete. The mixture was concentrated in vacuo to give 5- (1-methylcyclopropoxy) -3- [4- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] -2-pyridinyl ] -1H-indazole (200 mg, crude, HCl) as a yellow solid.
Step 7
To a mixture of 5- (1-methylcyclopropoxy) -3- [4- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] -2-pyridinyl ] -1H-indazole (78.47 mg,162.44umol,1 eq, HCl) and DIEA (41.99 mg,324.88umol,56.59ul,2 eq) in MeOH (5 mL) at 25 ℃ was added 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (60 mg,162.44umol,1 eq), CH3COOH (0.5 mL) and (2-methylpyridin-1-ium-1-yl) borohydride (52.12 mg,487.32umol,3 eq) at one time. The mixture was stirred at 25℃for 12 hours. The mixture was concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [2- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -4-pyridinyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (14.4 mg,16.68umol,10.27% yield, 98% purity, FA) as a yellow solid.
Exemplary Synthesis of Compound 99
Step 1
To a solution of 4-bromo-2, 5-difluoro-pyridine (300 mg,1.55mmol,729.93ul,1 eq.) in DMSO (5 mL) was added K2CO3 (427.50 mg,3.09mmol,2 eq.) and 4-piperidinylmethanol (267.19 mg,2.32mmol,1.5 eq.). The mixture was stirred at 90℃for 30min. TLC (petroleum ether: ethyl acetate=3:1, rf=0.24, uv 254 nm) showed the reaction was complete. The mixture was cooled to 25 ℃ and concentrated under reduced pressure at 25 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -40% (10 min) ethyl acetate/petroleum ether, 40% (10 min) ethyl acetate/petroleum ether) to give [1- (4-bromo-5-fluoro-2-pyridinyl) -4-piperidinyl ] methanol (500 mg, crude) as a yellow solid.
Step 2
To a mixture of [1- (4-bromo-5-fluoro-2-pyridinyl) -4-piperidinyl ] methanol (500 mg,933.79umol,54% purity, 1 eq.) in DCM (10 mL) was added DMP (792.12 mg,1.87mmol,578.19ul,2 eq.) at 25 ℃ in one portion. The mixture was stirred at 25℃for 30min. TLC (petroleum ether: ethyl acetate=1:1, rf=0.43, uv 254 nm) showed the reaction was complete. The residue was poured into NaHCO3 to adjust ph=7-8, and Na2SO3 (20 mL) was added. The aqueous phase was extracted with DCM (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (column height: 12g,100-200 mesh silica gel, 0% -10% (10 min) ethyl acetate/petroleum ether, 10% -20% (10 min) ethyl acetate/petroleum ether) to give 1- (4-bromo-5-fluoro-2-pyridinyl) piperidine-4-carbaldehyde (163 mg,533.63umol,57.15% yield, 94% purity) as a yellow oil.
Step 3
To a mixture of 1- (4-bromo-5-fluoro-2-pyridinyl) piperidine-4-carbaldehyde (163 mg,567.69umol,1 eq.) and tert-butyl piperazine-1-carboxylate (105.73 mg,567.69umol,1 eq.) in MeOH (10 mL) at 25 ℃ was added HOAc (1 mL) and borane in one portion; 2-methylpyridine (121.44 mg,1.14mmol,2 eq.). The mixture was stirred at 25℃for 16h. LCMS showed the desired MS. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,100-200 mesh silica gel, 0% -20% (15 min) ethyl acetate/petroleum ether, 20% (20 min) ethyl acetate/petroleum ether) to give tert-butyl 4- [ [1- (4-bromo-5-fluoro-2-pyridinyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (169 mg,369.50umol,65.09% yield) as a yellow oil.
Step 4
To a mixture of tert-butyl 4- [ [1- (4-bromo-5-fluoro-2-pyridinyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (169 mg,369.50umol,1 eq.), pin2B2 (187.66 mg,738.99umol,2 eq.) and KOAc (108.79 mg,1.11mmol,3 eq.) in dioxane (10 mL) was added Pd (dppf) Cl2 (13.52 mg,18.47umol,0.05 eq.) at 25℃in one portion. The mixture was stirred at 100℃for 1h. TLC showed the reaction was complete. The mixture was cooled to 25 ℃, filtered and concentrated in vacuo to give [2- [4- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] -1-piperidinyl ] -5-fluoro-4-pyridinyl ] boronic acid (430 mg,356.38umol,96.45% yield, 35% purity) as a black oil.
Step 5
To a mixture of [2- [4- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] -1-piperidinyl ] -5-fluoro-4-pyridinyl ] boronic acid (430 mg,356.38umol,35% purity, 1 eq), 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (174.21 mg,392.02umol,1.1 eq) and Na2CO3 (113.32 mg,1.07mmol,3 eq) in 1, 4-dioxane (10 mL) and H2O (2 mL) at 25 ℃ was added Pd (dppf) Cl2 (39.12 mg,53.46umol,0.15 eq) in one portion. The mixture was stirred at 100℃for 16 hours. LCMS showed the desired MS. The mixture was cooled to 25 ℃, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -10% ethyl acetate/petroleum ether) to give tert-butyl 4- [ [1- [ 5-fluoro-4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (76 mg,109.36umol,30.69% yield) as a yellow gum.
Step 6
To a mixture of tert-butyl 4- [ [1- [ 5-fluoro-4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (76 mg,109.36umol,1 eq.) in MeOH (5 mL) was added HCl/EtOAc (4 m,2mL,73.15 eq.) at 20 ℃ in one portion. The mixture was stirred at 65℃for 15min. LCMS showed the reaction was complete. The residue was concentrated in vacuo to give 3- [ 5-fluoro-2- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] -4-pyridinyl ] -5- (1-methylcyclopropoxy) -1H-indazole (51 mg,93.65umol,85.63% yield, 92% purity, HCl) as a yellow solid.
Step 7
To a mixture of 3- [ 5-fluoro-2- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] -4-pyridinyl ] -5- (1-methylcyclopropoxy) -1H-indazole (50 mg,107.62 mol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (39.75 mg,107.62 mol,1 eq) in MeOH (5 mL) at 25 ℃ was added borane in one portion; 2-methylpyridine (23.02 mg, 215.25. Mu.mol, 2 eq.) and HOAc (6.46 mg, 107.62. Mu.L, 6.16. Mu.L, 1 eq.). The mixture was stirred at 25 ℃ for 1h to give a yellow solution. LCMS showed about 57% of the required MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:10% -50%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [ 5-fluoro-4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (30.7 mg,35.71umol,33.18% yield, 95.13% purity) as a yellow solid.
Exemplary Synthesis of Compound 100
Compound 100 was prepared in a similar manner to compound 82 starting from tert-butyl 4- [4- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] oxy ] cyclohexyloxy ] piperidine-1-carboxylate.
Step 1
To a mixture of 4-hydroxycyclohexanone (30 g,262.83mmol,1 eq.) in pyridine (300 mL) was added dropwise 2, 2-dimethylpropionyl chloride (47.54 g,394.25mmol,48.51mL,1.5 eq.) at 0deg.C under N2. The mixture was stirred at 0 ℃ for 30min, then heated to 25 ℃ and stirred for 10 hours. TLC (petroleum ether: ethyl acetate=10:1, rf=0.72) showed that the reaction was complete. The residue was poured into water (200 mL). The aqueous phase was extracted with ethyl acetate (200 ml x 3). The combined organic phases were washed with brine (200 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (220 g,0% -7% (20 min) ethyl acetate/petroleum ether, 7% (30 min) ethyl acetate/petroleum ether) to give (4-oxocyclohexyl) 2, 2-dimethylpropionate (50 g,252.20mmol,95.95% yield) as a colorless oil.
Step 2
To a solution of 2, 2-dimethylpropionate (4-oxocyclohexyl) ester (50 g,252.20mmol,1 eq.) in EtOH (500 mL) at 0deg.C was added NaBH4 (10.04 g,265.40mmol,1.05 eq.). The mixture was then stirred at 0 ℃ for 1h to give a colorless solution. TLC (PE: etoac=3:1, rf=0.24) showed the reaction was complete. The mixture was poured into HCl (100 ml,2 m) and extracted with EtOAc (50 ml x 2). The organic layer was washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -30% (20 min) ethyl acetate/petroleum ether, 30% (30 min) ethyl acetate/petroleum ether) to give (4-hydroxycyclohexyl) 2, 2-dimethylpropionate (49 g,244.66mmol,97.01% yield) as a colorless oil.
Step 3
To a solution of 2, 2-dimethylpropionate (4-hydroxycyclohexyl) ester (49 g,244.66mmol,1 eq.) in THF (500 mL) at 0deg.C were added TEA (28.47 g,281.36mmol,39.16mL,1.15 eq.) and TMSCl (29.24 g,269.13mmol,34.16mL,1.1 eq.) and the reaction mixture was stirred at 25deg.C for 0.5 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. To a stirred solution of the above residue and benzyl 4-oxopiperidine-1-carboxylate (61.64 g,264.24mmol,52.68mL,1.08 eq.) in DCM (500 mL) was then added Et3SiH (32.72 g,281.36mmol,44.94mL,1.15 eq.) and TMSOTF (27.19 g,122.33mmol,22.11mL,0.5 eq.) dropwise at-60℃and the reaction mixture stirred under N2 for 1.5 h at 0 ℃. TLC (petroleum ether: ethyl acetate=3:1, rf=0.75, pma) showed the formation of new spots. The reaction mixture was quenched by addition of saturated NaHCO3 at 0 ℃ to adjust ph=8, followed by extraction with ethyl acetate (500 ml×3). The combined organic layers were washed with brine (500 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (0% -10% (10 min) ethyl acetate/petroleum ether, 10% (50 min) ethyl acetate/petroleum ether) to give benzyl 4- [4- (2, 2-dimethylpropionoyloxy) cyclohexyloxy ] piperidine-1-carboxylate (86.96 g,208.27mmol,85.12% yield) as a yellow gum.
Step 4
To a mixture of benzyl 4- [4- (2, 2-dimethylpropionoyloxy) cyclohexyloxy ] piperidine-1-carboxylate (50.00 g,119.75mmol,1 eq.) in EtOH (500 mL) at 25℃NaOH (23.95 g,598.75mmol,5 eq.) and H2O (150 mL) were added in one portion. The mixture was stirred at 80℃for 16 hours. TLC (PE: etoac=1:1) showed complete consumption of starting material and TLC showed a new spot. LCMS showed the desired MS. The mixture was cooled to 25 ℃. The residue was poured into water (500 mL). The aqueous phase was extracted with ethyl acetate (500 ml x 3). The combined organic phases were washed with brine (500 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -45% (20 min) ethyl acetate/petroleum ether, 45% (50 min) ethyl acetate/petroleum ether) to give benzyl 4- (((1 s,4 s) -4-hydroxycyclohexyl) oxy) piperidine-1-carboxylate (10 g,29.99mmol,25.05% yield) as a pale yellow gum and benzyl 4- (((1 r,4 r) -4-hydroxycyclohexyl) oxy) piperidine-1-carboxylate (3.68 g,11.04mmol,9.22% yield) as a pale yellow gum.
Step 5
To a solution of benzyl 4- (4-hydroxycyclohexyloxy) piperidine-1-carboxylate (1.50 g,4.50mmol,1 eq.) in THF (500 mL) at 0deg.C were added TEA (523.52 mg,5.17mmol,720.11uL,1.15 eq.) and TMSCL (537.63 mg,4.95mmol,628.07uL,1.1 eq.) and the reaction mixture was stirred at 25deg.C for 0.5 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. To a stirred solution of the above residue and benzyl 4-oxopiperidine-1-carboxylate (1.13 g,4.86mmol,968.94uL,1.08 eq.) in DCM (500 mL) was then added Et3SiH (601.59 mg,5.17mmol,826.36uL,1.15 eq.) and TMSOTF (499.95 mg,2.25mmol,406.47uL,0.5 eq.) dropwise at-60℃and the reaction mixture stirred under N2 at 0℃for 1.5 h. TLC (petroleum ether: ethyl acetate=0:1, rf=0.75, pma) showed the formation of new spots. LCMS showed the desired MS. The reaction mixture was quenched by addition of saturated NaHCO3 at 0 ℃ to adjust ph=8, followed by extraction with ethyl acetate (500 ml×3). The combined organic layers were washed with brine (500 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (0% -10% (10 min) ethyl acetate/petroleum ether, 10% (50 min) ethyl acetate/petroleum ether) to give benzyl 4- [4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] cyclohexyloxy ] piperidine-1-carboxylate (700 mg,1.27mmol,28.25% yield) as a yellow gum.
Step 6
To a solution of benzyl 4- [4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] cyclohexyloxy ] piperidine-1-carboxylate (700 mg,1.27mmol,1 eq.) in MeOH (50 mL) under N2 was added Pd/C (100 mg,1.27mmol,10% purity, 1 eq.). The suspension was degassed under vacuum and purged several times with H2. The mixture was stirred at 60℃under H2 (50 psi) for 16H. TLC (DCM/meoh=10/1, rf=0.51) indicated no residue of reactant 1 and a major new spot of greater polarity was detected. The reaction mixture was filtered, and the filtrate was concentrated to give 4- [4- (4-piperidinyloxy) cyclohexyloxy ] piperidine (510 mg, crude) as a white solid.
Step 7
To a solution of 4-bromo-2-fluoro-pyridine (96.59 mg,548.83 mol,0.5 eq) and 4- [4- (4-piperidinyloxy) cyclohexyloxy ] piperidine (310 mg,1.10mmol,1 eq) in DMSO (10 mL) was added DIEA (425.58 mg,3.29mmol,573.56uL,3 eq). The mixture was stirred at 100℃for 0.5h. LCMS showed the desired MS, boc2O (359.34 mg,1.65mmol,378.25ul,1.5 eq.) was added to the mixture and the mixture was stirred at 25 ℃ for 0.5h. LCMS showed about 29.8% of the required MS. The resulting product was poured into H2O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (0% -17% (10 min) ethyl acetate/petroleum ether, 17% (10 min) ethyl acetate/petroleum ether), LCMS showed about 47% of the desired MS. The crude product was purified by reverse phase HPLC (column Phenomenex Luna C18 75 x 30mm x 3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:45% -85%,35 min) to give tert-butyl 4- [4- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] oxy ] cyclohexyloxy ] piperidine-1-carboxylate (230 mg,369.65umol,33.68% yield, 86.55% purity) as a yellow solid.
Exemplary Synthesis of Compound 101
Step 1
To a solution of methyl 4-formylcyclohexane carboxylate (2 g,11.75mmol,1 eq.) and piperazine-1-carboxylate (2.85 g,12.93mmol,2.50mL,1.1 eq.) in MeOH (30 mL) and AcOH (3 mL) was added borane; 2-methylpyridine (6.28 g,58.75mmol,5 eq.). The mixture was stirred at 25℃for 4 hours. LC-MS (EB 134-923-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 89% of the desired compound was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent. The crude product was purified by column chromatography on silica gel (column height: 40g,100-200 mesh silica gel, 0% -100% (30 min) ethyl acetate/petroleum ether) to give benzyl 4- [ (4-methoxycarbonylcyclohexyl) methyl ] piperazine-1-carboxylate (4 g,10.68mmol,90.90% yield) as a yellow gum.
Step 2
DIBALH (1M, 5.34mL,2 eq.) was added dropwise to a solution of benzyl 4- [ (4-methoxycarbonylcyclohexyl) methyl ] piperazine-1-carboxylate (1 g,2.67mmol,1 eq.) in DCM (30 mL) at-78deg.C under N2 over a period of 30 min. During this time the temperature was kept below-78 ℃. The reaction mixture was warmed to 25 ℃ and stirred for 4 hours. LC-MS (EB 134-939-P1C) showed 27% reactant 1 remained. Several new peaks were shown on LC-MS and about 50% of the desired compound was detected. Saturated NH4Cl was added to the reaction mixture under ice-cooling. Saturated brine was added thereto, followed by extraction with ethyl acetate (50 ml×2). The organic layer was dried over anhydrous magnesium sulfate and concentrated. The crude product was purified by column chromatography on silica gel (column height: 40g,100-200 mesh silica gel, 0% -80% (15 min) ethyl acetate/petroleum ether) to give benzyl 4- [ [4- (hydroxymethyl) cyclohexyl ] methyl ] piperazine-1-carboxylate (310 mg,894.76umol,33.51% yield) as a colorless oil.
Step 3
To a solution of benzyl 4- [ [4- (hydroxymethyl) cyclohexyl ] methyl ] piperazine-1-carboxylate (310 mg,894.76umol,1 eq.) in DCM (6 mL) was added DMP (569.26 mg,1.34mmol,415.52uL,1.5 eq.). The mixture was stirred at 25℃for 2 hours. LC-MS (EB 134-941-P1B) showed no valid information. TLC (petroleum ether/ethyl acetate=1/2, rf=0.51) indicated no residue of reactant 1 and a major new spot of lower polarity was detected. The mixture was quenched with saturated NaHCO3 (30 mL x 3) and extracted with DCM (30 mL). The organic layer was washed with water (30 ml×2), brine (30 ml×2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give benzyl 4- [ (4-formylcyclohexyl) methyl ] piperazine-1-carboxylate as a yellow gum (300 mg,870.96umol,97.34% yield).
Step 4
To a solution of benzyl 4- [ (4-formylcyclohexyl) methyl ] piperazine-1-carboxylate (300 mg,870.96umol,1 eq.) and tert-butyl piperazine-1-carboxylate (243.33 mg,1.31mmol,1.5 eq.) in MeOH (6 mL) and AcOH (0.6 mL) was added borane; 2-methylpyridine (465.79 mg,4.35mmol,5 eq.). The mixture was stirred at 25℃for 4 hours. LC-MS (EB 134-943-P1B) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 42% of the desired compound was detected. The reaction mixture was diluted with water (20 mL) and extracted with EA (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by column chromatography on silica gel (column height: 8g,100-200 mesh silica gel, 0% -100% (30 min) ethyl acetate/petroleum ether) to give benzyl 4- [ [4- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] cyclohexyl ] methyl ] piperazine-1-carboxylate (350 mg,680.01umol,78.08% yield) as a yellow gum.
Step 5
To a solution of benzyl 4- [ [4- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] cyclohexyl ] methyl ] piperazine-1-carboxylate (350.00 mg,680.01umol,1 eq.) in EtOH (15 mL) was added Pd/C (60 mg,680.01umol,10% purity, 1 eq.) under N2. The suspension was degassed under vacuum and purged several times with H2. The mixture was stirred at 60℃under H2 (50 psi) for 16 hours. TLC (petroleum ether/ethyl acetate=0/1, rf=0) indicated no residue of reactant 1 and a major new spot of greater polarity was detected. The reaction mixture was filtered, and the filtrate was concentrated to give tert-butyl 4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ] methyl ] piperazine-1-carboxylate (210 mg,551.81umol,81.15% yield) as a white solid.
Step 6
To a solution of tert-butyl 4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ] methyl ] piperazine-1-carboxylate (210 mg,551.81umol,1 eq.) in DMSO (5 mL) was added DIEA (356.59 mg,2.76mmol,480.58uL,5 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (167.66 mg,606.99umol,1.1 eq.). The mixture was stirred at 100℃for 12 hours. LC-MS (EB 134-949-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 23.6% of the desired compound was detected. TLC (petroleum ether/ethyl acetate=0/1, rf=0.24) indicated that a major new spot of greater polarity was detected. The reaction mixture was diluted with water (20 mL) and extracted with EA (30 mL). The combined organic layers were washed with brine (20 ml x 3), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by column chromatography on silica gel (column height: 8g,100-200 mesh silica gel, 0% -80% (130 min) ethyl acetate/petroleum ether) to give tert-butyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] piperazin-1-yl ] methyl ] cyclohexyl ] methyl ] piperazine-1-carboxylate (170 mg,266.97umol,48.38% yield) as a yellow gum.
Step 7
To a solution of tert-butyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] cyclohexyl ] methyl ] piperazine-1-carboxylate (170 mg,266.97umol,1 eq.) in DCM (5 mL) was added TFA (1.54 g,13.51mmol,1mL,50.59 eq.). The mixture was stirred at 25℃for 2 hours. TLC (petroleum ether/ethyl acetate=0/1, rf=0) indicated no residue of reactant 1 and a major new spot of greater polarity was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (160 mg,230.16umol,86.21% yield, 93.6% purity, TFA) as a yellow gum.
Step 8
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (62.46 mg,116.38umol,1 eq.) in DMSO (5 mL) were added DIEA (150.41 mg,1.16mmol,202.71ul,10 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (35 mg,116.38umol,1 eq.). The mixture was stirred at 80℃for 12 hours. LC-MS (EB 134-962-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 80% of the desired compound was detected. The reaction mixture was diluted with water (20 mL) and extracted with EA (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The impure product was purified by preparative HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -30%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] cyclohexyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (48.3 mg,57.80umol,49.66% yield, 95.844% purity) as a yellow solid.
Exemplary Synthesis of Compound 102
Compound 101 was prepared in a similar manner to compound 82 using tert-butyl 4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ] methyl ] piperazine-1-carboxylate.
Exemplary Synthesis of Compound 103
Step 1
To a solution of methyl 2-oxo-1H-pyridine-4-carboxylate (15.00 g,97.95mmol,1 eq.) in MeOH (250 mL) under N2 was added Pd/C (6 g,10% purity, 1.00 eq.). The suspension was degassed under vacuum and purged several times with H2. The mixture was stirred at 25℃under H2 (15 psi) for 12 hours. TLC (DCM/meoh=10:1) showed complete consumption of starting material. The reaction mixture was filtered and the filtrate was concentrated to give methyl 2-oxopiperidine-4-carboxylate (16 g, crude) as a white solid.
Step 2
LAH (3.38 g,89.08mmol,2 eq.) was added in one portion to a mixture of methyl 2-oxopiperidine-4-carboxylate (7 g,44.54mmol,1 eq.) in THF (150 mL) at-40 ℃ under N2. The mixture was stirred at-40℃for 3 hours. TLC showed the reaction was complete. Water (5 mL) and 10% aqueous NaOH (5 mL) were added to the mixture at 0deg.C. The aqueous phase was dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 4- (hydroxymethyl) piperidin-2-one as a yellow oil (5.3 g,41.04mmol,92.13% yield).
Step 3
DMAP (501.32 mg,4.10mmol,0.1 eq.) and TosCl (15.65 g,82.07mmol,2 eq.) were added in portions to a mixture of 4- (hydroxymethyl) piperidin-2-one (5.3 g,41.04mmol,1 eq.) and TEA (12.46 g,123.11mmol,17.13mL,3 eq.) in DCM (120 mL) at 0deg.C. The mixture was stirred at 25℃for 12 hours. TLC showed the reaction was complete. The mixture was filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (DCM/meoh=100/1, 10/1) to give methyl 4-methylbenzenesulfonate (2-oxo-4-piperidinyl) ester (5.6 g,19.76mmol,48.16% yield) as a white solid.
Step 4
To a mixture of benzyl piperazine-1-carboxylate (466.43 mg,2.12mmol,409.15ul,1.5 eq.) and methyl 4-methylbenzenesulfonate (2-oxo-4-piperidinyl) ester (400 mg,1.41mmol,1 eq.) in MeCN (20 mL) at 25 ℃ was added DIEA (1.82 g,14.12mmol,2.46mL,10 eq.) in one portion. The mixture was stirred at 90℃for 12h, then (Boc) 2O (616.21 mg,2.82mmol,648.64uL,2 eq.) was added at 25℃and the mixture was stirred at 25℃for 2 h. LCMS showed the reaction was complete. The mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (DCM/meoh=100/1, 10/1) to give benzyl 4- [ (2-oxo-4-piperidinyl) methyl ] piperazine-1-carboxylate (370 mg,848.50umol,60.10% yield, 76% purity) as a yellow oil.
Step 5
To a mixture of benzyl 4- [ (2-oxo-4-piperidinyl) methyl ] piperazine-1-carboxylate (3 g,9.05mmol,1 eq.) in DMF (40 mL) at 0 ℃ was added NaH (724.11 mg,18.10mmol,60% purity, 2 eq.) in one portion. The mixture was stirred at 0deg.C for 30min, then tert-butyl 4- (p-toluenesulfonyloxymethyl) piperidine-1-carboxylate (4.01 g,10.86mmol,1.2 eq.) and KI (150.27 mg,905.23 mol,0.1 eq.) were added at 0deg.C and the mixture was stirred at 25deg.C for 11.5 h. TLC showed the reaction was complete. The mixture was poured into EtOAc (100 mL). The aqueous phase was washed with brine (100 ml x 3), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to give benzyl 4- [ [1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -2-oxo-4-piperidinyl ] methyl ] piperazine-1-carboxylate (500 mg,582.91umol,6.44% yield, 67% purity, FA) as a yellow solid.
Step 6
A mixture of 4- [ [1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -2-oxo-4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (400 mg,756.60 mol,1 eq.) was purified by SFC (column: DAICEL CHIRALCEL OJ (250 mm. Times.30 mm,10 um); mobile phase: [0.1% NH3H2O ETOH ]; B%:20% -20%, min) to give 4- [ [ (4R) -1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -2-oxo-4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester as a yellow oil or its enantiomer (180 mg,340.47 mol,45.00% yield) and 4- [ [ [ (4S) -1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -2-oxo-4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester as a yellow oil or its enantiomer (200 mg, 378.30.00% yield).
Step 7
To a solution of benzyl 4- [ [ (4R) -1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -2-oxo-4-piperidinyl ] methyl ] piperazine-1-carboxylate or its enantiomer (90 mg,170.23umol,1 eq.) in DCM (2 mL) was added TFA (1.54 g,13.51mmol,1mL,79.34 eq.). After the addition, the reaction solution was stirred at 20 ℃ for 1h. TLC showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove the solvent to give benzyl 4- [ [ (4R) -2-oxo-1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate or its enantiomer (72 mg, crude, TFA) as a yellow gum.
Step 8
To a solution of benzyl 4- [ [ (4R) -2-oxo-1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate or its enantiomer (72 mg,168.00umol,1.16 eq) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (40 mg,144.81umol,1 eq) in DMSO (3 mL) was added DIEA (56.15 mg,434.44umol,75.67ul,3 eq). After the addition, the mixture was stirred at 100 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -100% (20 min) ethyl acetate/petroleum ether, 0% -16% (10 min) dichloromethane/methanol, 16% (10 min) dichloromethane/methanol) to give 4- [ [ (4R) -1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] methyl ] -2-oxo-4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester, or enantiomer thereof (120 mg, crude material), as a yellow gum.
Step 9
To a mixture of benzyl 4- [ [ (4R) -1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -2-oxo-4-piperidinyl ] methyl ] piperazine-1-carboxylate or its enantiomer (120 mg,175.24umol,1 eq.) was added TFA (3.08 g,27.01mmol,2ml,154.14 eq.). The mixture was stirred at 80℃for 1h. TLC showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (4R) -2-oxo-4- (piperazin-1-ylmethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione or its enantiomer (96 mg,135.99umol,77.60% yield, 78% purity) as a yellow gum.
Step 10
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (4R) -2-oxo-4- (piperazin-1-ylmethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione or its enantiomer (96 mg,174.34umol,1.42 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (37 mg,123.03umol,1 eq.) in DMSO (2.5 mL) was added DIEA (371.00 mg,2.87mmol,0.5mL,23.33 eq.). After the addition, the mixture was stirred at 100 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (4R) -4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -2-oxo-1-piperidinyl ] isoindoline-1, 3-dione or enantiomer thereof (25.1 mg,30.34umol,24.66% yield, 98.5% purity) as a white solid.
Exemplary Synthesis of Compound 104
Compound 104 was prepared in a similar manner to compound 103 using benzyl 4- [ [ (4S) -1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -2-oxo-4-piperidinyl ] methyl ] piperazine-1-carboxylate or an enantiomer thereof.
Exemplary Synthesis of Compound 105
Step 1
To a solution of 4, 6-dibromopyrimidine (1 g,4.20mmol,1 eq) and tert-butyl 4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydro-2H-pyridine-1-carboxylate (1.56 g,5.04mmol,1.2 eq) in dioxane (20 mL) and H2O (2 mL) was added Na2CO3 (1.11 g,10.51mmol,2.5 eq) and Pd (dppf) Cl2 (153.80 mg,210.19umol,0.05 eq). After addition, the reaction mixture was stirred at 80 ℃ under N2 for 16h. LCMS showed the desired MS. TLC (petroleum ether=3:1) showed several spots. After cooling, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (30 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 30% ethyl acetate/petroleum ether) to give 4- (6-bromopyrimidin-4-yl) -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (650 mg,1.87mmol,44.54% yield, 98% purity) as a white solid.
Step 2
To a solution of trimethyl- [2- [ [5- (1-methylcyclopropoxy) -3-trimethylstannyl-indazol-1-yl ] methoxy ] ethyl ] silane (685 mg,1.42mmol,1.38 eq.) in THF (4 mL) was added tert-butyl 4- (6-bromopyrimidin-4-yl) -3, 6-dihydro-2H-pyridine-1-carboxylate (350 mg,1.03mmol,1 eq.), liCl (218.05 mg,5.14mmol,105.34ul,5 eq.), cuI (29.39 mg,154.31umol,0.15 eq.) and Pd (PPh 3) 4 (178.32 mg,154.31umol,0.15 eq.). After the addition, the reaction mixture was stirred by microwaves at 100 ℃ under N2 for 1.5h. LCMS showed the reaction was complete. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (3×10 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 25% ethyl acetate/petroleum ether) to give 4- [6- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (310 mg,480.19umol,46.68% yield, 89.5% purity) as a yellow gum.
Step 3
To a solution of 4- [6- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (310.00 mg,536.53umol,1 eq.) in MeOH (2 mL) was added Pd/C (100 mg,10% purity under Ar. After addition, the reaction mixture was degassed 3 times with H2 and stirred at 65 ℃ under H2 (50 psi) for 16H. TLC (petroleum ether: ethyl acetate=1:1) showed the reaction was complete. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 33% ethyl acetate/petroleum ether) to give 4- [6- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperidine-1-carboxylic acid tert-butyl ester as a yellow gum (150 mg,240.60umol,44.84% yield, 93% purity).
Step 4
To a solution of tert-butyl 4- [6- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperidine-1-carboxylate (150 mg,258.71umol,1 eq.) in MeOH (2 mL) was added HCl/EtOAc (4M, 2mL,30.92 eq.). After the addition, the reaction solution was stirred at 65 ℃ for 30min. LCMS showed the reaction was complete. After cooling, the reaction mixture was concentrated under reduced pressure. The resulting mixture was dissolved in DCM (5 mL) and DIEA (1 mL). The mixture was stirred at 25 ℃ for 5min and concentrated under reduced pressure to give 5- (1-methylcyclopropoxy) -3- [6- (4-piperidinyl) pyrimidin-4-yl ] -1H-indazole (90 mg, crude) as a brown solid. The crude product was used directly in the next step.
Step 5
To a solution of 5- (1-methylcyclopropoxy) -3- [6- (4-piperidinyl) pyrimidin-4-yl ] -1H-indazole (90 mg,257.56umol,1 eq.) and 4-formylpiperidine-1-carboxylic acid tert-butyl ester (109.86 mg,515.13umol,2 eq.) in MeOH (4 mL) and HOAc (0.4 mL) was added borane; 2-methylpyridine (55.10 mg,515.13umol,2 eq). After addition, the reaction was stirred at 20 ℃ for 12h. LCMS and TLC (pure ethyl acetate) showed the reaction was complete. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give tert-butyl 4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (120 mg,212.25umol,82.41% yield, 96.7% purity) as a pale yellow solid.
Step 6
To a solution of tert-butyl 4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (55 mg,100.60umol,1 eq.) in MeOH (2 mL) was added HCl/EtOAc (4M, 25.15uL,1 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 3h. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure. The resulting mixture was dissolved in DCM (5 mL) and DIEA (1 mL). The mixture was stirred at 25 ℃ for 5min and concentrated under reduced pressure to give 5- (1-methylcyclopropoxy) -3- [6- [1- (4-piperidinylmethyl) -4-piperidinyl ] pyrimidin-4-yl ] -1H-indazole (45 mg, crude) as a brown solid. The crude product was used directly in the next step.
Step 7
To a solution of 5- (1-methylcyclopropoxy) -3- [6- [1- (4-piperidinylmethyl) -4-piperidinyl ] pyrimidin-4-yl ] -1H-indazole (45.00 mg,100.76umol,1.01 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (37 mg,100.17umol,1 eq) in MeOH (5 mL) and HOAc (0.5 mL) was added borane; 2-methylpyridine (21.43 mg,200.34umol,2 eq). After the addition, the reaction solution was stirred at 20 ℃ for 12h. LCMS showed the reaction was complete. The mixture was diluted with water (10 mL) and extracted with dichloromethane (10 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (52.2 mg,65.11umol,65.00% yield, 99.78% purity) as a yellow solid.
Exemplary Synthesis of Compound 106
Compound 106 was prepared in a similar manner to compound 71 using 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione.
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (68.69 mg,120.79umol,1.1 eq, 3 TFA) and 3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) -1H-indazole (35 mg,109.81umol,1 eq) in DMSO (5 mL) at 20 ℃ was added DIEA portion (113.53 mg,878.48umol,153.01ul,8 eq). The mixture was stirred at 80℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow: 25mL/min; gradient time: 40min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 4-fluoro-4- [ [ (2S) -4- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -2-methyl-piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (20 mg,23.39umol,21.30% yield, 99.51% purity) as a yellow solid.
Exemplary Synthesis of Compound 107
Compound 107 was prepared in a similar manner to compound 73.
Step 1
To a mixture of 5- (1-methylcyclopropoxy) -3- [6- [4- [ [4- (4-piperidinylmethyl) piperazin-1-yl ] methyl ] -1-piperidinyl ] pyrimidin-4-yl ] -2H-indazole (50 mg,91.79 mol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5, 6-difluoro-isoindoline-1, 3-dione (30 mg,101.97 mol,1.11 eq.) in MeCN (5 mL) was added DIEA (371.00 mg,2.87mmol,0.5mL,31.27 eq.). The mixture was stirred at 60℃for 2h. LCMS showed the desired MS. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C1875*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow rate: 25mL/min; gradient time: 35min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-6- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (10.7 mg,12.94umol,14.09% yield, 99% purity) as a green solid.
Exemplary Synthesis of Compound 108
Compound 108 was prepared in a similar manner to compound 84 using trans-1, 4-bis (piperidin-4-yloxy) cyclohexane.
Step 1
DIEA (67.18 mg,519.83umol,90.54ul,8 eq.) was added in one portion to a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- (4-piperidinyloxy) cyclohexyloxy ] -1-piperidinyl ] isoindoline-1, 3-dione (35 mg,64.98umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (19.54 mg,64.98umol,1 eq.) in DMSO (5 mL) at 25 ℃. The mixture was stirred at 90℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 10% -50%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclohexyloxy ] -1-piperidinyl ] isoindoline-1, 3-dione (9.1 mg,11.23umol,17.29% yield, 99.10% purity) as a yellow solid.
Exemplary Synthesis of Compound 109
Step 1
To a mixture of DIEA (1.29 g,9.96mmol,1.73mL,2.42 eq.) and 3-aminopiperidine-2, 6-dione (812.61 mg,4.94mmol,1.2 eq., HCl) in DCM (30 mL) was added methyl 4-bromo-2-formyl-benzoate (1 g,4.11mmol,1 eq.) and AcOH (0.8 mL) at 25 ℃ in N2. The mixture was stirred at 40 ℃ for 12 hours, followed by addition of NaBH3CN (775.65 mg,12.34mmol,3 eq.) and stirring of the mixture at 40 ℃ for 4 hours. LCMS showed the reaction was complete. The mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 1/100) to give 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (500 mg,1.55mmol,37.61% yield) as a green solid.
Step 2
Cs2CO3 (231.91 mg,711.76umol,2 eq) and Pd-PEPPSI-pentcl-o-methylpyridine (19.32 mg,35.59umol,0.1 eq) were added at 25 ℃ in one portion to a mixture of 4- (dimethoxymethyl) piperidine (68.00 mg,427.06umol,1.2 eq) and 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (115 mg,355.88umol,1 eq) in DMSO (4 mL) at 25 ℃. The mixture was stirred at 80℃for 12 hours. The mixture was filtered and poured into ice saturated aqueous NH4Cl (30 mL). The aqueous phase was extracted with ethyl acetate (30 ml x 3). The combined organic phases were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 1/100) to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (90 mg,224.18umol,62.99% yield) as a yellow solid.
Step 3
To a mixture of 3- [5- [4- (dimethoxymethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (90 mg,224.18umol,1 eq.) in THF (4 mL) at 25 ℃ HCl (2 m,2mL,17.84 eq.) was added in one portion. The mixture was stirred at 25℃for 2 hours. LCMS showed the reaction was complete. The mixture was poured into saturated aqueous NaHCO3 (40 mL). The aqueous phase was extracted with ethyl acetate (40 ml x 3). The combined organic phases were washed with brine (40 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (60 mg,143.51umol,64.01% yield, 85% purity) as a yellow solid.
Step 4
To a mixture of 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (30 mg,84.42 mol,1 eq.) and 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (42.86 mg,92.86 mol,1.1 eq.) in DCM (2 mL) and MeOH (0.2 mL) was added HOAc (506.93 ug,8.44 mol,4.83e-1ul,0.1 eq.) at 25 ℃ in one portion. The mixture was stirred at 35 ℃ for 12 hours. NaBH3CN (15.91 mg,253.25umol,3 eq.) was then added and the mixture was stirred at 25℃for 4 hours. The mixture was concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,40 min) to give 3- [5- [4- [ [4- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (10.4 mg,12.87umol,15.24% yield, 99.11% purity) as a white solid.
Exemplary Synthesis of Compound 110
Compound 110 was prepared in a similar manner to compound 109.
Step 1
To a mixture of DIEA (20.60 mg,159.35 mol,27.76ul,2 eq) and 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1H-indazole (40 mg,79.68 mol,1 eq, HCl) in MeOH (10 mL) and HOAc (1 mL) at 25 ℃ was added 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (56.63 mg,159.35 mol,2 eq) and (2-methylpyridin-1-onium-1-yl) borohydride (25.57 mg,239.03 mol,3 eq) at one time. The mixture was stirred at 25℃for 16 hours. The mixture was concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex Luna C18 75 x 30mm x 3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:10% -40%,35 min) to give 3- [5- [4- [ [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] methyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (23.3 mg,28.37umol,35.60% yield, 98% purity) as a white solid.
Exemplary Synthesis of Compound 111
Step 1
To a mixture of tert-butyl (3S) -3- (hydroxymethyl) pyrrolidine-1-carboxylate (1 g,4.97mmol,1 eq.) and TosCl (1.89 g,9.94mmol,2 eq.) in DCM (10 mL) was added TEA (1.01 g,9.94mmol,1.38mL,2 eq.) and DMAP (607.01 mg,4.97mmol,1 eq.) at 0deg.C in N2 in one portion. The mixture was stirred at 20℃for 1 hour. LCMS showed the desired MS. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 50% ethyl acetate/petroleum ether) to give tert-butyl (3S) -3- (p-toluenesulfonyloxymethyl) pyrrolidine-1-carboxylate (2.2 g, crude) as a white solid.
Step 2
To a mixture of tert-butyl (3S) -3- (p-toluenesulfonyloxymethyl) pyrrolidine-1-carboxylate (462.53 mg,1.30mmol,1.5 eq.) and benzyl (3S) -3-methylpiperazine-1-carboxylate (500 mg,867.51umol,1 eq., 3 TFA) in MeCN (10 mL) at 20℃was added KI (720.04 mg,4.34mmol,5 eq.) and DIPEA (560.60 mg,4.34mmol,755.52uL,5 eq.) in one portion. The mixture was stirred at 100℃for 16 hours. TLC (DCM: meoh=10:1, rf=0.10) showed the reaction was complete. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (10 mL). The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ [ (3R) -1-tert-butoxycarbonylpyrrolidin-3-yl ] methyl ] -3-methyl-piperazine-1-carboxylate (460 mg, crude) as a yellow oil.
Step 3
To a mixture of benzyl (3S) -4- [ [ (3R) -1-tert-butoxycarbonylpyrrolidin-3-yl ] methyl ] -3-methyl-piperazine-1-carboxylate (460 mg,1.10mmol,1 eq.) in DCM (10 mL) at 25 ℃ was added TFA (1.54 g,13.51mmol,1mL,12.26 eq.) in one portion. The mixture was stirred at 25℃for 1 hour. LCMS showed the reaction was complete. The mixture was concentrated in vacuo to give benzyl (3S) -3-methyl-4- [ [ (3S) -pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate (500 mg, crude) as a yellow oil.
Step 4
To a mixture of benzyl (3S) -3-methyl-4- [ [ (3S) -pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate (350 mg,1.10mmol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (407.28 mg,1.10mmol,1 eq) in MeOH (10 mL) at 25 ℃ was added HOAc (1 mL) and borane in one portion; 2-methylpyridine (235.88 mg,2.21mmol,2 eq.). The mixture was stirred at 25℃for 1h. LCMS showed the desired MS. The mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10:1, 0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ [ (3S) -1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] methyl ] pyrrolidin-3-yl ] methyl ] -3-methyl-piperazine-1-carboxylate (1 g, crude) as a yellow oil.
Step 5
To a mixture of benzyl (3S) -4- [ [ (3S) -1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] pyrrolidin-3-yl ] methyl ] -3-methyl-piperazine-1-carboxylate (740 mg,1.10mmol,1 eq.) was added TFA (3.08 g,27.01mmol,2ml,24.49 eq.) at 70 ℃ in one portion. The mixture was stirred at 70℃for 3h. LCMS showed the desired MS. The residue was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (3S) -3- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] pyrrolidin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (1.2 g, crude) as a yellow oil.
Step 6
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (3S) -3- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] pyrrolidin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (182.02 mg,339.16umol,1.70 eq) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (60 mg,199.51umol,1 eq) in DMSO (5 mL) was added DIEA (206.28 mg,1.60mmol,278.00ul,8 eq) at 20 ℃. The mixture was stirred at 80℃for 12h. LCMS showed complete consumption of starting material. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow: 25mL/min; gradient time: 40min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (3S) -3- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] pyrrolidin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (62.6 mg,77.38umol,38.78% yield, 99% purity) as a yellow solid.
Exemplary Synthesis of Compound 112
Compound 112 was prepared in a similar manner to compound 109.
Step 1
A mixture of 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -2H-indazole (75.56 mg,168.83umol,1 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (60 mg,168.83umol,1 eq.) in HOAC (1 mL) and MeOH (10 mL) was stirred at 20deg.C for 20min, followed by the addition of borane; 2-methylpyridine (36.12 mg, 337.66. Mu. Mol,2 eq.). The mixture was then stirred at 20 ℃ under N2 for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -30%,40 min) to give 3- [5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (32.9 mg,41.81umol,24.76% yield, 100% purity) as a white solid.
Exemplary Synthesis of Compound 113
Compound 113 was prepared in a similar manner to compound 111 starting from tert-butyl (3R) -3- (hydroxymethyl) pyrrolidine-1-carboxylate.
Exemplary Synthesis of Compound 114
Step 1
To a mixture of tert-butyl (3S) -3- (p-toluenesulfonyloxymethyl) pyrrolidine-1-carboxylate (778.43 mg,2.19mmol,1.5 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-piperazin-1-yl-isoindoline-1, 3-dione (500 mg,1.46mmol,1 eq.) in MeCN (10 mL) was added KI (1.21 g,7.30mmol,5 eq.) and DIPEA (943.80 mg,7.30mmol,1.27mL,5 eq.) at 20℃in one portion. The mixture was stirred at 100℃for 16 hours. LCMS showed the desired product. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give (3R) -3- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] piperazin-1-yl ] methyl ] pyrrolidine-1-carboxylic acid tert-butyl ester (400 mg,738.21umol,50.56% yield, 97% purity) as a yellow oil.
Step 2
To a mixture of tert-butyl (3R) -3- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] pyrrolidine-1-carboxylate (400 mg,761.04umol,1 eq.) in DCM (5 mL) was added TFA (1.54 g,13.51mmol,1mL,17.75 eq.) at 20 ℃ in one portion. The mixture was stirred at 25℃for 1 hour. LCMS showed complete consumption of reaction 1 and found the desired MS. The mixture was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (3S) -pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (400 mg, crude) as a yellow oil.
Step 3
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (3S) -pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (323 mg,759.14umol,1 eq.) and tert-butyl 4-formylpiperidine-1-carboxylate (242.86 mg,1.14mmol,1.5 eq.) in MeOH (5 mL) at 25 ℃ was added HOAc (1 mL) and borane in one portion; 2-methylpyridine (162.40 mg,1.52mmol,2 eq.). The mixture was stirred at 25℃for 1h. LCMS showed the reaction was complete. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10:1, 0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 4- [ [ (3S) -3- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] piperazin-1-yl ] methyl ] pyrrolidin-1-yl ] methyl ] piperidine-1-carboxylate (600 mg, crude) as a yellow oil.
Step 4
To a mixture of tert-butyl 4- [ [ (3S) -3- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] pyrrolidin-1-yl ] methyl ] piperidine-1-carboxylate (600 mg,963.46umol,1 eq.) in DCM (10 mL) was added TFA (3.08 g,27.01mmol,2mL,28.04 eq.) at 25 ℃ in one portion. The mixture was stirred at 25℃for 1h. LCMS showed complete consumption of reaction 1 and found the desired MS. The mixture was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (3S) -1- (4-piperidinylmethyl) pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (1 g, crude TFA) as a yellow oil.
Step 5
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (3S) -1- (4-piperidinylmethyl) pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (177.26 mg,339.16 ul, 1.70 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (60 mg,199.51 ul, 1 eq.) in DMSO (5 mL) was added DIEA (206.28 mg,1.60mmol,278.00ul,8 eq.) at 20 ℃ in one portion. The mixture was stirred at 80℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow rate: 25mL/min; gradient time: 35min;100% B hold time: 3 min). The residue was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [ (3S) -1- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (38.2 mg,48.54umol,24.33% yield, 100% purity) as a yellow solid.
Exemplary Synthesis of Compound 115
Compound 115 was prepared in a similar manner to compound 114 starting from tert-butyl (3R) -3- (p-tolylsulfonyloxymethyl) pyrrolidine-1-carboxylate.
Exemplary Synthesis of Compound 116
Step 1
(3S) -3-hydroxypyrrolidine-1-carboxylic acid tert-butyl ester (5.00 g,26.70mmol,1 eq.) pyridin-4-ol (2.54 g,26.70mmol,1 eq.) and PPh3 (7.70 g,29.37mmol,1.1 eq.) were added to THF (50 mL) and stirred for 30 min. DIAD (5.94 g,29.37mmol,5.71mL,1.1 eq.) was added dropwise thereto at 0deg.C. Once the addition was complete, the reaction was stirred at 50 ℃ for 15.5 hours. TLC (dichloromethane: methanol=10:1, rf=0.45) showed the reaction was complete. LCMS showed the desired MS. The solvent was evaporated in vacuo. The resulting oil was treated with 1.0M aqueous HCl (100 mL). The acidic mixture was washed with CH2Cl2 (100 mL. Times.2). The combined CH2Cl2 washes were re-extracted with 1.0M aqueous HCl (100 mL) and H2O (200 mL) and then discarded. The aqueous fractions were combined, basified to pH about 12 using 1.0M aqueous NaOH, and extracted with CH2Cl2 (50 ml×4). The organic extracts were washed with brine, dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (40 g,100-200 mesh silica gel, 0% -100% (20 min) ethyl acetate/petroleum ether) to give tert-butyl (3R) -3- (4-pyridyloxy) pyrrolidine-1-carboxylate (1.8 g,6.81mmol,25.50% yield) as a yellow oil.
Step 2
To a solution of tert-butyl (3R) -3- (4-pyridyloxy) pyrrolidine-1-carboxylate (1.8 g,6.81mmol,1 eq.) in EtOH (20 mL) and HOAc (408.94 mg,6.81mmol,389.46uL,1 eq.) was added Pt2O (257.73 mg,6.81mmol,1 eq.) at 25 ℃. The mixture was then stirred at 70℃under H2 (50 psi) for 16H. LCMS showed the desired MS. The suspension was filtered through a pad of celite and the pad or filter cake was washed with EtOH (100 mL x 3). The combined filtrates were concentrated to dryness to give tert-butyl (3R) -3- (4-piperidinyloxy) pyrrolidine-1-carboxylate (1.5 g,5.55mmol,81.47% yield) as a colorless oil.
Step 3
To a mixture of tert-butyl 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (100 mg,232.02umol,1 eq), (3R) -3- (4-piperidinyloxy) pyrrolidine-1-carboxylate (150 mg,554.80umol,2.39 eq) in DMSO (5 mL) was added Et3N (70.43 mg,696.05umol,96.88ul,3 eq) at once followed by stirring at 100 ℃ for 1h. TLC showed complete consumption of starting material. The mixture was cooled to 20 ℃, then the residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (3X 5 mL). The combined organic phases were washed with brine (2×5 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -5% (5 min) ethyl acetate/petroleum ether, 5% (10 min) ethyl acetate/petroleum ether, 5% -30% (5 min) ethyl acetate/petroleum ether, 30% (5 min) ethyl acetate/petroleum ether) to give (3R) -3- [ [1- [6- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] pyrrolidine-1-carboxylic acid tert-butyl ester (100 mg,147.09umol,63.40% yield, 97.8% purity) as a yellow oil.
Step 4
To a mixture of tert-butyl (3R) -3- [ [1- [6- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] pyrrolidine-1-carboxylate (100.00 mg,150.40 mol,1 eq.) in MeOH (5 mL) was added HCl/EtOAc (4M, 112.80ul,3 eq.) in one portion at 25 ℃. The mixture was stirred at 65℃for 0.5h. TLC (petroleum ether: ethyl acetate=5:1, rf=0.03) showed that the reaction was complete. The mixture was concentrated in vacuo to give 5- (1-methylcyclopropoxy) -3- [6- [4- [ (3R) -pyrrolidin-3-yl ] oxy-1-piperidinyl ] pyrimidin-4-yl ] -1H-indazole (80 mg,118.90umol,79.06% yield, 70% purity, HCl) as a yellow solid.
Step 5
To a mixture of 5- (1-methylcyclopropoxy) -3- [6- [4- [ (3R) -pyrrolidin-3-yl ] oxy-1-piperidinyl ] pyrimidin-4-yl ] -1H-indazole (80 mg,128.87umol,70% purity, 1 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (47.60 mg,128.87umol,1 eq.) in MeOH (20 mL) at 25 ℃ was added borane in one portion; 2-methylpyridine (27.57 mg,257.75umol,2 eq.) and HOAc (1 mL). The mixture was stirred at 25℃for 16h. LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (3R) -3- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] pyrrolidin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (45.4 mg,57.19umol,44.38% yield, 99.25% purity) as a yellow solid.
Exemplary Synthesis of Compound 117
Compound 117 was prepared in a similar manner to compound 116 starting from tert-butyl (3R) -3-hydroxypyrrolidine-1-carboxylate.
Exemplary Synthesis of Compound 118
Step 1
To a solution of tert-butyl (3S) -3- (4-piperidinyloxy) pyrrolidine-1-carboxylate (734.05 mg,2.72mmol,1.5 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (500 mg,1.81mmol,1 eq.) in DMSO (10 mL) and DIEA (2.34 g,18.10mmol,3.15mL,10 eq.). The mixture was stirred at 80℃for 16h. LCMS showed the desired product MS. The resulting product was poured into H2O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (0% -50% (10 min) ethyl acetate/petroleum ether, 50% (10 min) ethyl acetate/petroleum ether) to give (3S) -3- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] pyrrolidine-1-carboxylic acid tert-butyl ester (760 mg,1.44mmol,79.74% yield) as a yellow solid.
Step 2
To a mixture of tert-butyl (3S) -3- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] pyrrolidine-1-carboxylate (300 mg,569.71umol,1 eq.) in DCM (5 mL) was added TFA (3.08 g,27.01mmol,2mL,47.41 eq.) at 25 ℃ in one portion. The mixture was stirred at 25℃for 0.5h. TLC (petroleum ether: ethyl acetate=5:1, rf=0.03) showed that the reaction was complete. The mixture was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ (3S) -pyrrolidin-3-yl ] oxy-1-piperidinyl ] isoindoline-1, 3-dione (300 mg,438.49umol,76.97% yield, 79% purity, TFA) as a yellow solid.
Step 3
To a mixture of 2- (2, 6-dioxopiperidin-3-yl) -5- (4- ((S) -pyrrolidin-3-yloxy) piperidin-1-yl) isoindoline-1, 3-dione (242 mg,567.46 mol,1 eq.) and tert-butyl 4-formylpiperidine-1-carboxylate (181.53 mg,851.18 mol,1.5 eq.) in MeOH (10 mL) at 25 ℃ was added HOAc (1 mL) and borane in one portion; 2-methylpyridine (121.39 mg,1.13mmol,2 eq.). The mixture was stirred at 25℃for 1h. TLC (dichloromethane: methanol=10:1, rf=0.50) showed the reaction was complete. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10:1, rf=0.50, 0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 4- (((3S) -3- ((1- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl) piperidin-4-yl) oxy) pyrrolidin-1-yl) methyl) piperidine-1-carboxylate (270 mg,388.72umol,68.50% yield, 89.8% purity) as a yellow oil.
Step 4
To a mixture of tert-butyl 4- [ [ (3S) -3- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] pyrrolidin-1-yl ] methyl ] piperidine-1-carboxylate (120 mg,192.39umol,1 eq.) in DCM (5 mL) was added TFA (3.08 g,27.01mmol,2mL,140.41 eq.) at 25 ℃ in one portion. The mixture was stirred at 25℃for 0.5h. TLC (petroleum ether: ethyl acetate=5:1, rf=0.03) showed that the reaction was complete. The mixture was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ (3S) -1- (4-piperidinylmethyl) pyrrolidin-3-yl ] oxy-1-piperidinyl ] isoindoline-1, 3-dione (100 mg,148.99umol,77.44% yield, 95% purity, TFA) as a yellow gum.
Step 5
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ (3S) -1- (4-piperidinylmethyl) pyrrolidin-3-yl ] oxy-1-piperidinyl ] isoindoline-1, 3-dione (100 mg,156.83 mol,1.57 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (30 mg,99.75 mol,1 eq) in DMSO (5 mL) was added DIEA (103.14 mg,798.03 mol,139.00ul,8 eq) at once at 25 ℃. The mixture was stirred at 90℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ (3S) -1- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] pyrrolidin-3-yl ] oxy-1-piperidinyl ] isoindoline-1, 3-dione (21.5 mg,26.91umol,26.98% yield, 98.63% purity) as a yellow solid.
Exemplary Synthesis of Compound 119
Compound 119 was prepared in a similar manner to compound 118 using (3S) -3- (4-piperidinyloxy) pyrrolidine-1-carboxylic acid tert-butyl ester.
Exemplary Synthesis of Compound 120
Step 1
To a solution of methyl 5-oxopyrrolidine-3-carboxylate (10 g,69.86mmol,1 eq.) in THF (200 mL) at 0 ℃ under N2 was added LiAlH4 (3.98 g,104.79mmol,1.5 eq.) in portions. After the addition, the reaction mixture was stirred at 0 ℃ for 1h. TLC (ethyl acetate: methanol=10:1) showed several new spots. The reaction mixture was quenched by the addition of H2O (2 mL), followed by 15% aqueous NaOH (2 mL) and water (6 mL). After stirring at room temperature for 30min, the mixture was filtered through a pad of celite to remove solids. The filtrate was concentrated to dryness to give the crude product. The residue was purified by silica gel column chromatography (0% to 10% methanol/ethyl acetate) to give 4- (hydroxymethyl) pyrrolidin-2-one (7.84 g, crude) as a white solid.
Step 2
To a mixture of 4- (hydroxymethyl) pyrrolidin-2-one (390 mg,3.39mmol,1 eq.) and TosCl (1.29 g,6.77mmol,2 eq.) in DCM (10 mL) was added TEA (685.56 mg,6.77mmol,942.99ul,2 eq.) and DMAP (206.92 mg,1.69mmol,0.5 eq.) at 0 ℃ in one portion under N2. The mixture was stirred at 20℃for 1 hour. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=0:1) showed several new spots. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give methyl 4-methylbenzenesulfonate (5-oxopyrrolidin-3-yl) ester (550 mg,1.67mmol,49.44% yield, 82% purity) as a brown solid.
Step 3
To a solution of 4-methylbenzenesulfonic acid (5-oxopyrrolidin-3-yl) methyl ester (7 g,25.99mmol,1 eq.) in MeCN (200 mL) was added benzyl piperazine-1-carboxylate (6.87 g,31.19mmol,6.03mL,1.2 eq.), KI (12.94 g,77.98mmol,3 eq.) and Cs2CO3 (21.17 g,64.98mmol,2.5 eq.). The mixture was then stirred at 80℃for 16 hours. TLC (dichloromethane: methanol=10:1, rf=0.4) showed no starting material and a new spot. The residue was diluted with H2O (50 mL) and extracted with ethyl acetate (100 mL. Times.3). The combined organic layers were washed with brine (2×50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% -10% methanol/dichloromethane) to give benzyl 4- [ (5-oxopyrrolidin-3-yl) methyl ] piperazine-1-carboxylate (4.2 g,11.65mmol,44.80% yield, 88% purity) as a colorless gum.
Step 4
A solution of NaH (1.06 g,26.47mmol,60% purity, 3 eq.) in DMF (10 mL) was cooled to 0deg.C, after which 4- [ (5-oxopyrrolidin-3-yl) methyl ] piperazine-1-carboxylic acid benzyl ester (2.8 g,8.82mmol,1 eq.) was slowly added. The solution was stirred at 0deg.C for another 10 minutes, and tert-butyl 4- (p-toluenesulfonyloxymethyl) piperidine-1-carboxylate (4.24 g,11.47mmol,1.3 eq.) and KI (4.39 g,26.47mmol,3 eq.) were added. The reaction mixture was stirred at 25 ℃ for 16 hours. LCMS showed the desired MS. The resulting product was poured into H2O (10 mL). The mixture was extracted with ethyl acetate (50 ml x 3). The organic phase was washed with brine (50 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to give benzyl 4- [ [1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -5-oxo-pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate (450 mg,874.37umol,9.91% yield) as a colorless oil.
Step 5
4- [ [1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -5-oxo-pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylic acid benzyl ester (450 mg,874.37umol,1 eq.) was purified by SFC. The residue was purified by SFC (column: DAICEL CHIRALPAK AD (250 mm. Times.30 mm,10 um); mobile phase: [0.1% NH3H2O IPA ]; B%:25% -25%, min) to give benzyl 4- [ [ (3R) -1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -5-oxo-pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate or its enantiomer (160 mg,310.89umol,35.56% yield, 100% purity) and benzyl 4- [ [ [ (3S) -1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -5-oxo-pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate or its enantiomer (160 mg,296.90umol,33.96% yield, 95.5% purity) as a colorless oil.
Step 6
To a mixture of benzyl 4- [ [ (3R) -1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -5-oxo-pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate or its enantiomer (50 mg,97.15umol,1 eq.) in DCM (3 mL) was added TFA (11.08 mg,97.15umol,7.19ul,1 eq.) followed by stirring the mixture at 25 ℃ for 1 hour. TLC (dichloromethane: methanol=10:1) showed a new spot. The resulting product was concentrated in vacuo to give benzyl 4- [ [ (3R) -5-oxo-1- (4-piperidinylmethyl) pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate or enantiomer thereof (50 mg, crude, TFA) as a residue as a colorless oil.
Step 7
To a mixture of benzyl 4- [ [ (3R) -5-oxo-1- (4-piperidinylmethyl) pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate or its enantiomer (40.27 mg,97.15umol,1 eq), 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (26.83 mg,97.15umol,1 eq) in DMSO (5 mL) was added DIEA (12.56 mg,97.15umol,16.92ul,1 eq), and the mixture was stirred at 100 ℃ under an atmosphere of N2 for 12 hours. LCMS showed the desired MS. TLC (methanol: dichloromethane=1:10) showed a new major spot. The resulting product was poured into H2O (10 mL). The mixture was extracted with ethyl acetate (50 ml x 3). The organic phase was washed with brine (50 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (0% to 10% methanol/dichloromethane) to give 4- [ [ (3R) -1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -5-oxo-pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylic acid benzyl ester or enantiomer thereof (100 mg,73.05umol,75.20% yield, 49% purity) as a yellow oil.
Step 8
To a mixture of 4- [ [ (3R) -1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -5-oxo-pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylic acid benzyl ester or its enantiomer (65 mg,96.91umol,1 eq.) in TFA (11.05 mg,96.91umol,7.18ul,1 eq.) the mixture was then stirred at 70 ℃ under an atmosphere of N2 for 1 hour. TLC (dichloromethane: methanol=10:1) showed complete consumption of starting material and a new spot was found. The reaction mixture was filtered and concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (4R) -2-oxo-4- (piperazin-1-ylmethyl) pyrrolidin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione or its enantiomer (70 mg, crude TFA) as a colorless oil.
Step 9
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (4R) -2-oxo-4- (piperazin-1-ylmethyl) pyrrolidin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione or its enantiomer (53.53 mg,99.75 mol,1 eq), 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (30 mg,99.75 mol,1 eq), DIEA (12.89 mg,99.75 mol,17.38ul,1 eq) in DMSO (5 mL) was added DIEA (12.89 mg,99.75 mol,17.38ul,1 eq) followed by stirring the mixture under an N2 atmosphere at 100 ℃ for 12 hours. LCMS showed complete consumption of starting material and found the desired MS. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to afford 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [ (4R) -4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -2-oxo-pyrrolidin-1-yl ] methyl ] -1-piperidyl ] isoindoline-1, 3-dione or enantiomer thereof (11.5 mg,13.78 mol,13.82% yield, 96% purity) as a yellow solid.
Exemplary Synthesis of Compound 121
Compound 121 was prepared in a similar manner to compound 120 using benzyl 4- [ [ (3S) -1- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] -5-oxo-pyrrolidin-3-yl ] methyl ] piperazine-1-carboxylate or an enantiomer thereof.
Exemplary Synthesis of Compound 122
Step 1
To a mixture of tert-butyl 3- (hydroxymethyl) azetidine-1-carboxylate (500 mg,2.67mmol,1 eq.) and TosCl (1.02 g,5.34mmol,2 eq.) in DCM (10 mL) were added TEA (270.22 mg,2.67mmol,371.69ul,1 eq.) and DMAP (326.25 mg,2.67mmol,1 eq.) at 0 ℃ in one portion. The mixture was stirred at 20 ℃ for 1 hour to give a pale yellow mixture. TLC (petroleum ether: ethyl acetate=1:1, rf=0.30) showed that the reaction was complete. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -22% (5 min) ethyl acetate/petroleum ether, 22% (5 min) ethyl acetate/petroleum ether) to give tert-butyl 3- (p-toluenesulfonyloxymethyl) azetidine-1-carboxylate (1 g, crude) as a colorless oil.
Step 2
To a mixture of tert-butyl 3- (p-tolylsulfonyloxymethyl) azetidine-1-carboxylate (444.28 mg,1.30mmol,1.5 eq.) and benzyl (3S) -3-methylpiperazine-1-carboxylate (500 mg,867.51umol,1 eq., 3 TFA) in MeCN (5 mL) at 20℃was added KI (720.03 mg,4.34mmol,5 eq.) and DIPEA (560.58 mg,4.34mmol,755.50uL,5 eq.) in one portion. The mixture was stirred at 100℃for 16 hours. TLC showed the reaction was complete. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 3), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,30ml/min,0% -10% (10 min) MeOH/DCM,10% (10 min) MeOH/DCM) to give benzyl (3S) -4- [ (1-tert-butoxycarbonylazetidin-3-yl) methyl ] -3-methyl-piperazine-1-carboxylate (170 mg,311.76umol,35.94% yield, 74% purity) as a yellow gum.
Step 3
To a mixture of benzyl (S) -4- ((1- (tert-butoxycarbonyl) azetidin-3-yl) methyl) -3-methylpiperazine-1-carboxylate (170 mg,421.30 mol,1 eq) in DCM (5 mL) was added TFA (4.62 g,40.52mmol,3mL,96.17 eq) at 25 ℃ in one portion. The mixture was stirred at 25℃for 30min. TLC showed the reaction was complete. The mixture was concentrated in vacuo to give benzyl (S) -4- (azetidin-3-ylmethyl) -3-methylpiperazine-1-carboxylate (200 mg,244.36umol,58.00% yield, 51% purity, TFA) as a yellow gum.
Step 4
To a mixture of benzyl (3S) -4- (azetidin-3-ylmethyl) -3-methyl-piperazine-1-carboxylate (200 mg,479.13umol,1 eq, TFA) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (176.98 mg,479.13umol,1 eq) in MeOH (10 mL) at 25 ℃ was added HOAc (1 mL) in one portion; 2-methylpyridine (102.50 mg,958.26umol,2 eq.). The mixture was stirred at 25℃for 1h. TLC (dichloromethane: methanol=10:1, rf=0.59) showed the reaction was complete. LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10:1, rf= 0.43,0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give benzyl (3S) -4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] methyl ] azetidin-3-yl ] methyl ] -3-methyl-piperazine-1-carboxylate (150 mg,212.40umol,44.33% yield, 93% purity) as a yellow oil.
Step 5
A mixture of (3S) -4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] azetidin-3-yl ] methyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (150 mg,228.39umol,1 eq.) in TFA (3.34 g,29.30mmol,2.17mL,128.28 eq.) at 20℃in one portion. The mixture was stirred at 70 ℃ for 2 hours to give a yellow solution. LCMS showed the desired MS. The residue was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] azetidin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione as a yellow gum (150 mg,149.86umol,65.62% yield, 75% purity, 2 TFA).
Step 6
DIEA (206.27 mg,1.60mmol,278.00uL,8 eq.) was added at 20deg.C in N2 to a mixture of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [3- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] azetidin-1-yl ] methyl ] -1-piperidyl ] isoindoline-1, 3-dione (150 mg,199.82 uL, 1.00 eq., 2 TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (60 mg,199.51 uL, 1 eq.) in DMSO (5 mL). The mixture was stirred at 80℃for 36h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow: 25mL/min; gradient time: 40min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] azetidin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (13 mg,15.85umol,7.94% yield, 95.95% purity) as a yellow solid.
Exemplary Synthesis of Compound 123
Compound 123 was prepared in a similar manner to compound 122 starting from tert-butyl 3-fluoro-3- (hydroxymethyl) azetidine-1-carboxylate.
Exemplary Synthesis of Compound 124
Step 1
To a solution of methyl 3-formylbicyclo [1.1.1] pentane-1-carboxylate (250 mg,1.62mmol,1 eq.) and piperazine-1-carboxylate (535.80 mg,2.43mmol,470.00ul,1.5 eq.) in MeOH (5 mL) and HOAc (0.5 mL) was added borane; 2-methylpyridine (520.36 mg,4.86mmol,3 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 1h. LCMS showed the desired MS. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 50% ethyl acetate/petroleum ether) to give benzyl 4- [ (3-methoxycarbonyl-1-bicyclo [1.1.1] pentyl) methyl ] piperazine-1-carboxylate (370 mg,1.03mmol,63.66% yield) as a colorless oil.
Step 2
To a solution of benzyl 4- [ (3-methoxycarbonyl-1-bicyclo [1.1.1] pentyl) methyl ] piperazine-1-carboxylate (370 mg,1.03mmol,1 eq.) in DCM (5 mL) at-78 ℃ was added DIBAL-H (1 m,3.10mL,3 eq.) and the resulting mixture was stirred under N2 at-78 ℃ for 1H. The reaction was then warmed to 25 ℃ for 12h. TLC (petroleum ether: ethyl acetate=1:1) showed several new spots. The reaction mixture was quenched with NH4Cl (15 mL) and filtered. The resulting filtrate was extracted with ethyl acetate (50 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 70% ethyl acetate/petroleum ether) to give benzyl 4- [ [3- (hydroxymethyl) -1-bicyclo [1.1.1] pentyl ] methyl ] piperazine-1-carboxylate (120 mg,337.75umol,32.72% yield, 93% purity) as a pale yellow oil.
Step 3
To a solution of benzyl 4- [ [3- (hydroxymethyl) -1-bicyclo [1.1.1] pentyl ] methyl ] piperazine-1-carboxylate (120 mg,363.17 mol,1 eq.) in DCM (2 mL) was added DMP (308.07 mg,726.35 mol,224.87uL,2 eq.). After the addition, the reaction mixture was stirred at 25 ℃ for 1h. TLC (petroleum ether: ethyl acetate=1:2) showed the reaction was complete. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give benzyl 4- [ (3-formyl-1-bicyclo [1.1.1] pentyl) methyl ] piperazine-1-carboxylate (120 mg, crude) as a yellow gum. The crude product was used directly in the next step.
Step 4
To a solution of benzyl 4- [ (3-formyl-1-bicyclo [1.1.1] pentyl) methyl ] piperazine-1-carboxylate (120 mg,365.40umol,1 eq.) and tert-butyl piperazine-1-carboxylate (136.11 mg,730.81umol,2 eq.) in MeOH (3 mL) and HOAc (0.3 mL) was added borane; 2-methylpyridine (195.42 mg,1.83mmol,5 eq.). After addition, the reaction mixture was stirred at 25 ℃ for 12h. TLC (pure ethyl acetate) showed the reaction was complete and several new spots formed. The reaction mixture was neutralized to pH 7 with saturated NaHCO 3. The resulting mixture was extracted with ethyl acetate (10 ml x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% -100% ethyl acetate/petroleum ether to 0% -20% methanol/dichloromethane) to give benzyl 4- [ [3- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] -1-bicyclo [1.1.1] pentyl ] methyl ] piperazine-1-carboxylate (100 mg,200.54umol,54.88% yield, 100% purity) as a pale yellow oil.
Step 5
To a solution of benzyl 4- [ [3- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] -1-bicyclo [1.1.1] pentyl ] methyl ] piperazine-1-carboxylate (100 mg,200.54 mol,1 eq.) in DCM (2 mL) was added TFA (1.08 g,9.45mmol,0.7mL,47.14 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 2 hours. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give benzyl 4- [ [3- (piperazin-1-ylmethyl) -1-bicyclo [1.1.1] pentyl ] methyl ] piperazine-1-carboxylate (100 mg, crude, TFA) as a yellow gum. The crude product was used directly in the next step.
Step 6
To a solution of benzyl 4- [ [3- (piperazin-1-ylmethyl) -1-bicyclo [1.1.1] pentyl ] methyl ] piperazine-1-carboxylate (100 mg,195.10umol,1 eq., TFA) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (53.89 mg,195.10umol,1 eq.) in DMSO (2 mL) was added DIEA (176.51 mg,1.37mmol,237.88ul,7 eq.). After the addition, the reaction solution was stirred at 100 ℃ for 12 hours. LCMS showed the reaction was complete. After cooling, the reaction mixture was diluted with ethyl acetate (20 mL) and washed with brine (20 mL x 2). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative TLC (dichloromethane: methanol=10:1) to give benzyl 4- [ [3- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -1-bicyclo [1.1.1] pentyl ] methyl ] piperazine-1-carboxylate (50 mg,75.60umol,38.75% yield, 99% purity) as a yellow solid.
Step 7
A mixture of 4- [ [3- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -1-bicyclo [1.1.1] pentyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (50 mg,76.36umol,1 eq) and TFA (2 mL) was stirred at 80℃for 1h. TLC (dichloromethane: methanol=10:1) showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- (piperazin-1-ylmethyl) -1-bicyclo [1.1.1] pentyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (40 mg, crude) as a yellow gum. The crude product was used directly in the next step.
Step 8
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- (piperazin-1-ylmethyl) -1-bicyclo [1.1.1] pentyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (40.00 mg,76.83 mol,1.00 eq.) in DMSO (2 mL) was added 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (23 mg,76.48 mol,1 eq.) and DIEA (59.31 mg,458.86 mol,79.93uL,6 eq.). After the addition, the reaction solution was stirred at 90 ℃ for 12 hours. LCMS showed about 37% of the desired MS. After cooling, the reaction mixture was diluted with ethyl acetate (20 mL) and washed with brine (20 mL x 2). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-bicyclo [1.1.1] pentyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (18.3 mg,22.99umol,30.06% yield, 98.6% purity) as a yellow solid.
Exemplary Synthesis of Compound 125
Step 1
To a mixture of 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (200 mg,665.02umol,1 eq.) and 4- (dimethoxymethyl) piperidine (158.83 mg,997.53umol,1.5 eq.) in DMSO (5 mL) was added DIEA (5.94 g,45.93mmol,8.00mL,69.06 eq.) and the mixture was stirred at 80℃under N2 for 2 hours. LCMS showed complete consumption of starting material and found the desired MS. TLC (petroleum ether: ethyl acetate=1:1) showed a new main point. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (0% to 50% petroleum/ethyl acetate) to give 3- [6- [4- (dimethoxymethyl) -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1H-indazole (268 mg,601.17umol,90.40% yield, 95% purity) as a yellow solid.
Step 2
To a mixture of 3- [6- [4- (dimethoxymethyl) -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1H-indazole (140 mg,330.57 mol,1 eq) in THF (2 mL) was added HCl (2 m,5.09mL,30.80 eq) and the mixture was stirred at 80 ℃ under N2 for 2 hours. LCMS showed complete consumption of starting material and found the desired MS. The residue was poured into NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (140 mg,259.64umol,78.54% yield, 70% purity) as a white solid.
Step 3
To a solution of 1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (60 mg,158.97umol,1 eq) and 5- [4- (azetidin-3-ylmethyl) piperazin-1-yl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (80 mg,194.43umol,1.22 eq) in MeOH (10 mL) and HOAc (1 mL) was added borane; 2-methylpyridine (45 mg,420.71umol,2.65 eq). After the addition, the reaction solution was stirred at 25 ℃ for 3h. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] azetidin-3-yl ] methyl ] piperazin-1, 3-dione (68.3 mg,87.57umol,55.09% yield, 99.10% purity) as a yellow solid.
Exemplary Synthesis of Compound 126
Step 1
To a solution of tert-butyl 4- (3-hydroxycyclobutoxy) piperidine-1-carboxylate (3 g,11.06mmol,1 eq) and pyridin-4-ol (1.58 g,16.58mmol,1.5 eq) in THF (150 mL) was added PPh3 (4.35 g,16.58mmol,1.5 eq) and the mixture was stirred at 25 ℃ for 0.5 h. And DIAD (3.35 g,16.58mmol,3.22mL,1.5 eq.) was added to the above mixture at 0deg.C under N2, followed by stirring the mixture at 90deg.C under N2 for 16 hours. TLC (pure ethyl acetate) showed the reaction was complete. After cooling, the reaction mixture was concentrated under reduced pressure. The resultant was diluted with 1M HCl (50 mL) and washed with ethyl acetate (50 mL x 2). The aqueous phase was neutralized to pH about 7 with 1M NaOH and extracted with ethyl acetate (40 ml x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: phenomenex luna C, 250, 80mm, 10um; mobile phase: [ water (0.225% FA) -ACN ]; B%:20% -50%,15 min) to give tert-butyl 4- [3- (4-pyridyloxy) cyclobutoxy ] piperidine-1-carboxylate (2.7 g,6.66mmol,60.28% yield, 86% purity) as a pale yellow oil.
Step 2
To a solution of tert-butyl 4- [3- (4-pyridyloxy) cyclobutoxy ] piperidine-1-carboxylate (2.7 g,7.75mmol,1 eq.) in toluene (50 mL) was added BnBr (1.33 g,7.75mmol,920.37uL,1 eq.). After addition, the reaction was stirred at 80 ℃ for 12h. After cooling, the reaction mixture was concentrated under reduced pressure. The residue was dissolved in MeOH (50 mL) and washed with petroleum ether (50 mL x 3). The MeOH phase was concentrated under reduced pressure to give tert-butyl 4- [3- (1-benzyl-pyridin-1-ium-4-yl) oxy-cyclobutoxy ] piperidine-1-carboxylate (3.69 g, crude) as a pale yellow gum.
Step 3
To a solution of tert-butyl 4- [3- (1-benzylpyridin-1-ium-4-yl) oxybutyloxy ] piperidine-1-carboxylate (3.69 g,8.39mmol,1 eq.) in EtOH (50 mL) was added NaBH4 (1.020g, 26.96mmol,3.21 eq.) at 0deg.C. After the addition, the reaction solution was stirred at 25 ℃ for 12h. TLC (petroleum ether: ethyl acetate=5:1) and LCMS showed the reaction was complete. The reaction mixture was diluted with brine (60 mL) and extracted with ethyl acetate (60 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 40% ethyl acetate/petroleum ether) to give tert-butyl 4- [3- [ (1-benzyl-3, 6-dihydro-2H-pyridin-4-yl) oxy ] cyclobutoxy ] piperidine-1-carboxylate (1.8 g,3.90mmol,46.51% yield, 96% purity) as a pale yellow oil.
Step 4
To a solution of tert-butyl 4- [3- [ (1-benzyl-3, 6-dihydro-2H-pyridin-4-yl) oxy ] cyclobutoxy ] piperidine-1-carboxylate (1.8 g,4.07mmol,1 eq.) in MeOH (20 mL) at 25℃was added Pd/C (0.2 g,10% purity) and Pd (OH) 2 (0.2 g,142.41umol,10% purity). The suspension was degassed under vacuum and purged several times with H2. The mixture was then stirred at 60℃under H2 (50 psi) for 16H. LCMS showed the reaction was complete. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 4- [3- (4-piperidinyloxy) cyclobutoxy ] piperidine-1-carboxylate (1.42 g,4.01mmol,98.50% yield) as a pale red oil.
Step 5
To a solution of tert-butyl 4- [3- (4-piperidinyloxy) cyclobutoxy ] piperidine-1-carboxylate (200 mg,564.20umol,1 eq) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (155.84 mg,564.20umol,1 eq) in DMSO (5 mL) was added DIEA (218.76 mg,1.69mmol,294.82uL,3 eq). After the addition, the reaction mixture was stirred at 100 ℃ for 12 hours. LCMS showed the starting material was consumed and the desired MS formed. After cooling, the reaction mixture was diluted with ethyl acetate (20 mL) and washed with water (20 mL x 2). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 75% ethyl acetate/petroleum ether) to give tert-butyl 4- [3- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (300 mg,382.68umol,67.83% yield, 77.9% purity) as a yellow solid.
Step 6
To a solution of tert-butyl 4- [3- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (300 mg,491.24umol,1 eq.) in EtOAc (3 mL) was added HCl/EtOAc (4M, 3mL,24.43 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 1h. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [3- (4-piperidinyloxy) cyclobutoxy ] -1-piperidinyl ] isoindoline-1, 3-dione (260 mg,384.98umol,78.37% yield, 81% purity, HCl) as a yellow solid. The crude product was used directly in the next step.
Step 7
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [3- (4-piperidinyloxy) cyclobutoxy ] -1-piperidinyl ] isoindoline-1, 3-dione (84.89 mg,166.26umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (50 mg,166.26umol,1 eq.) in DMSO (2 mL) was added DIEA (107.44 mg,831.28umol,144.79uL,5 eq.). After the addition, the reaction solution was stirred at 90 ℃ for 12h. LCMS showed the reaction was complete. After cooling, the reaction solution was diluted with water (10 mL) and extracted with dichloromethane (10 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 10% -50%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [3- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclobutoxy ] -1-piperidinyl ] isoindoline-1, 3-dione (37.7 mg,48.02umol,28.88% yield, 98.7% purity) as a yellow solid.
Exemplary Synthesis of Compound 127
Step 1
To a solution of tert-butyl 4- [3- (4-piperidinyloxy) cyclobutoxy ] piperidine-1-carboxylate (320 mg,902.72umol,1 eq.) and 4-bromo-2-fluoro-pyridine (320 mg,1.82mmol,2.01 eq.) in CH3CN (5 mL) was added Cs2CO3 (588.25 mg,1.81mmol,2 eq.). After the addition, the reaction mixture was stirred at 80 ℃ for 12 hours. LCMS showed the reaction was complete. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 50% ethyl acetate/petroleum ether) to give tert-butyl 4- [3- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (240 mg,446.65umol,49.48% yield, 95% purity) as a white solid.
Step 2
KOAc (92.29 mg,940.32umol,2 eq) and Pd (dppf) Cl2 (17.20 mg,23.51umol,0.05 eq) were added in one portion to a mixture of tert-butyl 4- [3- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (240 mg,470.16umol,1 eq) and Pin2B2 (238.78 mg,940.32umol,2 eq) in dioxane (5 mL) at 25 ℃. The mixture was stirred at 100℃for 16h. LCMS showed the desired MS. The reaction mixture was concentrated under reduced pressure to remove the solvent to give tert-butyl 4- [3- [ [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (500 mg, crude) as a black oil.
Step 3
To a solution of tert-butyl 4- [3- [ [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (262.00 mg,469.93umol,2.09 eq.) and Na2CO3 (71.55 mg,675.10umol,3 eq.) in dioxane (5 mL) and H2O (1 mL) was added 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (100 mg,225.03umol,1 eq.), pd (dppf) Cl2 (8.23 mg,11.25umol,0.05 eq.). After addition, the reaction mixture was stirred at 90 ℃ under N2 for 16h. LCMS showed the desired MS. After cooling, the reaction mixture was filtered to remove insoluble materials, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (12 g,0% -30% (8 min) ethyl acetate/petroleum ether, 30% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 4- [3- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (200 mg, crude) as a yellow gum.
Step 4
To a mixture of tert-butyl 4- [3- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (200 mg,267.37umol,1 eq.) in MeOH (3 mL) was added HCl/dioxane (4 m,3mL,44.88 eq.). The mixture was stirred at 65℃for 30min. TLC showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 5- (1-methylcyclopropoxy) -3- [2- [4- [3- (4-piperidinyloxy) cyclobutoxy ] -1-piperidinyl ] -4-pyridinyl ] -1H-indazole (138 mg, crude, HCl) as a yellow solid.
Step 5
To a solution of 5- (1-methylcyclopropoxy) -3- [2- [4- [3- (4-piperidinyloxy) cyclobutoxy ] -1-piperidinyl ] -4-pyridinyl ] -1H-indazole (138 mg,266.58 mol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (73.64 mg,266.58 mol,1 eq.) in DMSO (3 mL) was added DIEA (172.27 mg,1.33mmol,232.17uL,5 eq.). After the addition, the mixture was stirred at 100 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 10% -60%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [3- [ [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] oxy ] cyclobutoxy ] -1-piperidinyl ] isoindoline-1, 3-dione (21.2 mg,26.71umol,10.02% yield, 97.5% purity) as a yellow solid.
Exemplary Synthesis of Compound 128
Compound 128 was prepared in a similar manner to compound 78 starting with azetidin-3-yl-methanol.
Exemplary Synthesis of Compound 129
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindoline-1, 3-dione (250 mg,568.82umol,1 eq.), tert-butyl 3- (p-toluenesulfonyloxymethyl) azetidine-1-carboxylate (233.05 mg,682.58umol,1.2 eq.) and KI (472.12 mg,2.84mmol,5 eq.) in MeCN (5 mL) was added DIEA (367.57 mg,2.84mmol,495.38ul,5 eq.) at 25 ℃. The mixture was stirred at 100℃for 12 hours. TLC (dichloromethane: methanol=10/1) showed the reaction was complete. The mixture was cooled to 25 ℃ and concentrated under reduced pressure. The residue was purified by silica gel chromatography (dichloromethane/methanol=10/1) to give tert-butyl 3- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] azetidine-1-carboxylate (300 mg,482.98umol,84.91% yield, 98% purity) as a yellow solid.
Step 2
To a mixture of tert-butyl 3- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] azetidine-1-carboxylate (150 mg,246.42umol,1 eq.) in DCM (2 mL) was added TFA (1.54 g,13.51mmol,1mL,54.81 eq.) in one portion. The mixture was stirred at 25℃for 1 hour. TLC (dichloromethane: methanol=10/1) showed the reaction was complete. The mixture was concentrated under reduced pressure at 35 ℃. The crude product was used in the next step without further purification. The product 5- [4- [ [4- (azetidin-3-ylmethyl) piperazin-1-yl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (100 mg,188.75umol,76.60% yield, 96% purity) was a pale yellow oil.
Step 3
To a mixture of 5- [4- [ [4- (azetidin-3-ylmethyl) piperazin-1-yl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (100 mg,196.61umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (59.13 mg,196.61umol,1 eq.) in DMSO (2 mL) was added DIEA (254.11 mg,1.97mmol,342.47ul,10 eq.) in one portion. The mixture was stirred at 84℃for 3 hours. LCMS showed that reactant EB153-509-P1 remained 9% and yielded 51% of the desired product. The mixture was cooled to 25 ℃ and poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 2). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex Luna C, 75X 30mm X3 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,35 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] azetidin-3-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (45.7 mg,55.53umol,28.24% yield, 99.5% purity, FA) as a yellow solid.
Exemplary Synthesis of Compound 130
Step 1
Tert-butyl 3-hydroxyazetidine-1-carboxylate (5 g,28.87mmol,1 eq.) pyridin-4-ol (2.75 g,28.87mmol,1 eq.) and PPh3 (8.33 g,31.75mmol,1.1 eq.) were added to THF (50 mL) and stirred for 30 minutes. DIAD (6.42 g,31.75mmol,6.17mL,1.1 eq.) was added dropwise thereto at 0deg.C. Once the addition was complete, the reaction was stirred at 50 ℃ for 15.5 hours. TLC (dichloromethane: methanol=10:1, rf=0.45) showed the reaction was complete. LCMS showed the desired MS. The solvent was evaporated in vacuo. The resulting oil was treated with 1.0M aqueous HCl (100 mL). The acidic mixture was washed with CH2Cl2 (100 mL. Times.2). The combined CH2Cl2 washes were re-extracted with 1.0M aqueous HCl (100 mL) and H2O (200 mL) and then discarded. The aqueous fractions were combined, basified to pH about 12 using 1.0M aqueous NaOH, and extracted with CH2Cl2 (50 ml×4). The organic extracts were washed with brine, dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (40 g,100-200 mesh silica gel, 0% -100% (20 min) ethyl acetate/petroleum ether) to give tert-butyl 3- (4-pyridyloxy) azetidine-1-carboxylate (1.8 g,7.19mmol,24.91% yield) as a yellow oil.
Step 2
To a solution of tert-butyl 3- (4-pyridyloxy) azetidine-1-carboxylate (1.8 g,7.19mmol,1 eq.) in EtOH (20 mL) and HOAc (431.85 mg,7.19mmol,411.29uL,1 eq.) was added Pt2O (257.73 mg,7.19mmol,1 eq.) at 25 ℃. The mixture was then stirred at 70℃under H2 (50 psi) for 16H. TLC showed the reaction was complete. The suspension was filtered through a pad of celite and the pad or filter cake was washed with EtOH (100 mL x 3). The combined filtrates were concentrated to dryness to give tert-butyl 3- (4-piperidinyloxy) azetidine-1-carboxylate (2.1 g, crude) as a colorless oil.
Step 3
To a mixture of tert-butyl 3- (4-piperidinyloxy) azetidine-1-carboxylate (208.20 mg,812.19umol,2 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (150 mg,406.10umol,1 eq.) in MeOH (5 mL) at 20 ℃ was added borane in one portion; 2-methylpyridine (86.87 mg, 812.19. Mu. Mol,2 eq.) and HOAc (0.5 mL). The mixture was stirred at 20℃for 1h to give a yellow solution. LCMS showed about 50% of the required MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -100% (15 min) ethyl acetate/petroleum ether, 100% (15 min) ethyl acetate/petroleum ether) to give tert-butyl 3- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -4-piperidinyl ] oxy ] azetidine-1-carboxylate (130 mg,213.22umol,52.50% yield) as a yellow solid.
Step 4
To a mixture of tert-butyl 3- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -4-piperidinyl ] oxy ] azetidine-1-carboxylate (130 mg,213.22umol,1 eq.) in DCM (5 mL) was added TFA (3.41 g,29.94mmol,2.22mL,140.41 eq.) in one portion at 20 ℃. The mixture was stirred at 20℃for 0.5h. TLC (dichloromethane: methanol=10:1, rf=0.03) showed the reaction was complete. The mixture was concentrated in vacuo to give 5- [4- [ [4- (azetidin-3-yloxy) -1-piperidinyl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (120 mg,169.33umol,79.42% yield, 88% purity, TFA) as a yellow gum.
Step 5
To a mixture of 5- [4- [ [4- (azetidin-3-yloxy) -1-piperidinyl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (120 mg,192.43umol,1.16 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (50 mg,166.26umol,1 eq) in DMSO (5 mL) at 20 ℃ was added DIEA (1.48 g,11.48mmol,2mL,69.07 eq) in one portion. The mixture was stirred at 80℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] azetidin-3-yl ] oxy-1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (72.1 mg,93.06umol,55.98% yield, 99.89% purity) as a yellow solid.
Exemplary Synthesis of Compound 131
Step 1
To a mixture of tert-butyl 3-fluoro-3- (hydroxymethyl) azetidine-1-carboxylate (500 mg,2.44mmol,1 eq.) and TEA (739.60 mg,7.31mmol,1.02mL,3 eq.) in DCM (10 mL) was added TosCl (696.73 mg,3.65mmol,1.5 eq.) and DMAP (29.76 mg,243.63umol,0.1 eq.) at 0deg.C in N2. The mixture was stirred at 25℃for 12 hours. TLC showed the reaction was complete. The mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 1/1) to give tert-butyl 3-fluoro-3- (p-tolylsulfonyloxymethyl) azetidine-1-carboxylate (800 mg,2.09mmol,85.88% yield, 94% purity) as a yellow solid.
Step 2
To a mixture of benzyl piperazine-1-carboxylate (520.93 mg,2.36mmol,456.95ul,1 eq.) and tert-butyl 3-fluoro-3- (p-tolylsulfonyloxymethyl) azetidine-1-carboxylate (850 mg,2.36mmol,1 eq.) in DMSO (3 mL) at 25℃was added DIEA (1.53 g,11.82mmol,2.06mL,5 eq.) in one portion. The mixture was stirred at 90℃for 12 hours. TLC showed formation of the desired product. The residue was poured into ethyl acetate (30 mL). The combined organic phases were washed with brine (30 ml x 3), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 1/1) to give benzyl 4- [ (1-tert-butoxycarbonyl-3-fluoro-azetidin-3-yl) methyl ] piperazine-1-carboxylate (900 mg,2.21mmol,93.39% yield) as a yellow oil.
Step 3
To a mixture of benzyl 4- [ (1-tert-butoxycarbonyl-3-fluoro-azetidin-3-yl) methyl ] piperazine-1-carboxylate (200 mg,490.82umol,1 eq) in DCM (4 mL) at 25℃was added TFA (1.54 g,13.51mmol,1mL,27.52 eq) in one portion. The mixture was stirred at 25℃for 2 hours. TLC showed the reaction was complete. The mixture was concentrated in vacuo to give benzyl 4- [ (3-fluoroazetidin-3-yl) methyl ] piperazine-1-carboxylate (200 mg,365.46umol,74.46% yield, 77% purity, TFA) as a yellow solid.
Step 4
To a mixture of benzyl 4- [ (3-fluoroazetidin-3-yl) methyl ] piperazine-1-carboxylate (200 mg,474.63umol,1 eq, TFA) and 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (409.13 mg,474.63umol,50% purity, 1 eq) in DMSO (5 mL) was added DIEA (613.43 mg,4.75mmol,826.72ul,10 eq) at 25 ℃ in one portion. The mixture was stirred at 90℃for 12 hours. TLC showed the reaction was complete. The mixture was poured into ethyl acetate (20 mL). The combined organic phases were washed with brine (20 ml x 3), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 1/1) to give benzyl 4- [ [ 3-fluoro-1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] azetidin-3-yl ] methyl ] piperazine-1-carboxylate (300 mg,414.59umol,87.35% yield, 97% purity) as a yellow solid.
Step 5
A mixture of benzyl 4- [ [ 3-fluoro-1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] azetidin-3-yl ] methyl ] piperazine-1-carboxylate (70 mg,99.73umol,1 eq.) in TFA (3 mL) was stirred at 70℃for 1 hour. TLC showed the reaction was complete. The mixture was concentrated under reduced pressure. The residue was poured into THF (5 mL) and NH3H2O (1 mL). The mixture was stirred at 25 ℃ for 20min, dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 3- [6- [ 3-fluoro-3- (piperazin-1-ylmethyl) azetidin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (50 mg,51.67umol,51.81% yield, 57% purity, TFA) as a yellow solid.
Step 6
To a mixture of 3- [6- [ 3-fluoro-3- (piperazin-1-ylmethyl) azetidin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (50 mg,90.66umol,1 eq, TFA) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (33.49 mg,90.66umol,1 eq) in MeOH (10 mL) and CH3COOH (1 mL) at 25 ℃ was added in one portion (29.09 mg,271.97umol,3 eq). The mixture was stirred at 25℃for 12 hours. TLC showed the reaction was complete. The mixture was concentrated in vacuo. The residue was purified by preparative HPLC (column 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35% for 40min; and column 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (10 mM NH4HCO 3) -ACN ]; B%:20% -70%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ 3-fluoro-1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] azetidin-3-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (14.4 mg,18.03umol,19.88% yield, 99% purity) as a yellow solid.
Exemplary Synthesis of Compound 132
Compound 132 was prepared in a similar manner to compound 78 starting from benzyl 4- [ (3-fluoroazetidin-3-yl) methyl ] piperazine-1-carboxylate.
Exemplary Synthesis of Compound 133
Step 1
To a solution of tert-butyl 3- (hydroxymethyl) azetidine-1-carboxylate (1 g,5.34mmol,1 eq.) in DCM (3 mL) was added TFA (4.62 g,40.52mmol,3mL,7.59 eq.). The mixture was stirred at 25℃for 1 hour. LC-MS (EB 2049-122-P1A) showed complete consumption of reactant 1 and the required mass was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent to give azetidin-3-yl-methanol (1 g,4.97mmol,93.09% yield, TFA) as a pale yellow oil. The crude product was used in the next step without further purification.
Step 2
To a solution of azetidin-3-ylmethanol (1 g,4.97mmol,1 eq, TFA) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (1.37 g,4.97mmol,1 eq) in DMSO (6 mL) was added DIEA (3.21 g,24.86mmol,4.33mL,5 eq). The mixture was stirred at 100℃for 16 hours. LC-MS (EB 2049-123-P1A) showed complete consumption of reactant 1 and the required mass was detected. The reaction mixture was diluted with water (20 mL) and extracted with EA (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel 12g/>Silica gel flash column, eluent: 0% -50% ethyl acetate/Petroleum ether gradient, 45 mL/min) to give 2- (2, 6-dioxo-3-piperidyl) -5- [3- (hydroxymethyl) azetidin-1-yl as a yellow solid]Isoindoline-1, 3-dione (520 mg,1.51mmol,30.46% yield).
Step 3
To 2- (2, 6-dioxo-3-piperidyl)) -5- [3- (hydroxymethyl) azetidin-1-yl]To a solution of isoindoline-1, 3-dione (500 mg,1.46mmol,1 eq.) in DCM (5 mL) was added DMAP (35.58 mg, 291.26. Mu. Mol,0.2 eq.), toscl (555.29 mg,2.91mmol,2 eq.) and TEA (294.73 mg,2.91mmol, 405.40. Mu.L, 2 eq.). The mixture was stirred at 25℃for 16 hours. LC-MS (EB 2049-124-P1A) showed complete consumption of reactant 1 and the required mass was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent. The residue was purified by flash chromatography on silica gel12g/>Silica gel flash column, eluent: 0% -100% ethyl acetate/petroleum ether gradient, 45 mL/min) to afford 4-methylbenzenesulfonic acid [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] as a yellow solid]Azetidin-3-yl]Methyl ester (186 mg,373.85umol,25.67% yield).
Step 4
To tert-butyl 4- (4-piperidinylmethyl) piperazine-1-carboxylate (98.63 mg,348.03umol,1.5 eq.) and 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl]Methoxy group]To a solution of ethyl-trimethyl-silane (100 mg, 232.02. Mu.L, 1 eq.) in DMSO (3 mL) was added DIEA (89.96 mg, 696.05. Mu.L, 121.24. Mu.L, 3 eq.). The mixture was stirred at 100℃for 1 hour. LC-MS (EB 2049-126-P1B) showed complete consumption of reactant 1 and the desired mass was detected. The mixture was cooled to room temperature and concentrated, then the residue was quenched with saturated NaHCO3 (30 mL) and extracted with EtOAc (30 mL). The organic layer was washed with water (30 ml×2), brine (30 ml×2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by flash chromatography on silica gel12gSilica gel flash column, eluent: 0% -35% ethyl acetate/petroleum ether gradient, 45 mL/min) to afford 4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] as a pale yellow oil]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Tert-butyl piperazine-1-carboxylate (107 mg,157.83umol,68.02% yield).
Step 5
To a solution of tert-butyl 4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (107 mg,157.83 mol,1 eq.) in MeOH (3 mL) was added HCl/dioxane (4 m,39.46ul,1 eq.). The mixture was stirred at 65℃for 0.5 h. LC-MS (EB 2049-128-P1A) showed complete consumption of reactant 1 and detection of a desired mass. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -2H-indazole (80 mg,139.87umol,88.62% yield, 91% purity, 2 HCl) as a pale yellow oil. The crude product was used in the next step without further purification.
Step 6
To a solution of 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -2H-indazole (80 mg,165.28 mmol,1 eq., HCl) and 4-methylbenzenesulfonic acid [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] azetidin-3-yl ] methyl ester (82.23 mg,165.28 mmol,1 eq.) in MeCN (3 mL) was added KI (274.36 mg,1.65mmol,10 eq.) and DIEA (213.61 mg,1.65mmol,287.88uL,10 eq.). The mixture was stirred at 100℃for 16 hours. LC-MS (EB 2049-129-P1C) showed complete consumption of reactant 1 and the required mass was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [3- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] azetidin-1, 3-dione (26.6 mg,33.73umol,20.41% yield, 98% purity) as a yellow solid.
Exemplary Synthesis of Compound 134
Step 1
To a mixture of 1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (100 mg,264.94umol,1 eq) and 3-fluoro-3- (piperazin-1-ylmethyl) azetidine-1-carboxylic acid tert-butyl ester (72.42 mg,264.94umol,1 eq) in MeOH (10 mL) and CH3COOH (1 mL) was added at 25 ℃ in one portion (2-methylpyridin-1-ium-1-yl) borohydride (85.02 mg,794.83umol,3 eq). The mixture was stirred at 25℃for 12 hours. TLC showed the reaction was complete. The mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (DCM/meoh=100/1, 10/1) to give 3-fluoro-3- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] azetidine-1-carboxylic acid tert-butyl ester (110 mg,86.64umol,32.70% yield, 50% purity) as a yellow solid.
Step 2
To a mixture of tert-butyl 3-fluoro-3- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] azetidine-1-carboxylate (100 mg,78.77umol,50% purity, 1 eq.) in DCM (4 mL) was added TFA (1.54 g,13.51mmol,1mL,171.47 eq.) at 25 ℃ in one portion. The mixture was stirred at 25℃for 2 hours. TLC showed the reaction was complete. The mixture was concentrated in vacuo to give 3- [6- [4- [ [4- [ (3-fluoroazetidin-3-yl) methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (100 mg, crude TFA) as a yellow oil.
Step 3
To a mixture of 3- [6- [4- [ [4- [ (3-fluoroazetidin-3-yl) methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (100 mg,77.08umol,50% purity, 1 eq, TFA) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (27.68 mg,100.20umol,1.3 eq) in DMSO (3 mL) was added DIEA (99.62 mg,770.78umol,134.26ul,10 eq) at 25 ℃. The mixture was stirred at 100℃for 12 hours. TLC showed the reaction was complete. The mixture was concentrated in vacuo. The mixture was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (10 mM NH4HCO 3) -ACN ]; B%:30% -80%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [ 3-fluoro-3- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] azetidin-1-yl ] isoindoline-1, 3-dione (10.6 mg,13.13umol,17.04% yield, 98% purity) as a yellow solid.
Exemplary Synthesis of Compound 135
Step 1
To a solution of tert-butyl 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carboxylate (139.86 mg,464.03umol,2 eq.) and 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (100 mg,232.02umol,1 eq.) in DMSO (3 mL) was added DIEA (371.00 mg,2.87mmol,0.5mL,12.37 eq.). After the addition, the reaction solution was stirred at 100 ℃ for 2h. LCMS showed the desired MS. The residue was poured into water (15 mL). The aqueous phase was extracted with ethyl acetate (15 ml x 3). The combined organic phases were washed with brine (15 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -37% (10 min) ethyl acetate/petroleum ether, 37% (10 min) ethyl acetate/petroleum ether) to give tert-butyl 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] ] piperidine-1-carboxylate (140 mg,181.05umol,78.03% yield, 90% purity) as a yellow oil.
Step 2
To a mixture of tert-butyl 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (140 mg,201.17umol,1 eq.) in MeOH (2 mL) was added HCl/dioxane (4 m,2mL,39.77 eq.). The mixture was stirred at 65℃for 30min. LCMS showed the desired MS. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (93 mg, crude, HCl) as a white solid.
Step 3
To a solution of 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (93.66 mg,201.17 mol,1 eq.) and 4-methylbenzenesulfonic acid [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] azetidin-3-yl ] methyl ester (100.09 mg,201.17 mol,1 eq.) in MeCN (3 mL) were added DIEA (130.00 mg,1.01mmol,175.20uL,5 eq.) and KI (166.97 mg,1.01mmol,5 eq.). After the addition, the reaction solution mixture was stirred at 80 ℃ for 16h. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [3- [ [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] methyl ] azetidin-1-yl ] isoindoline-1, 3-dione (43.8 mg,53.17umol,26.43% yield, 96% purity) as a yellow solid.
Exemplary Synthesis of Compound 136
Step 1
To a mixture of 1-tert-butoxycarbonylazetidine-3-carboxylic acid (273.56 mg,1.36mmol,1.2 eq.) and HATU (516.93 mg,1.36mmol,1.2 eq.) in DCM (4 mL) was added DIEA (732.11 mg,5.66mmol,986.67ul,5 eq.) at 25 ℃ in one portion, the mixture was stirred at 25 ℃ for 10min, followed by benzyl 4- [ (4-fluoro-4-piperidinyl) methyl ] piperazine-1-carboxylate (380 mg,1.13mmol,1 eq.). The mixture was stirred at 25℃for 120min. TLC (petroleum ether: ethyl acetate=0/1) showed that new spots formed and LCMS showed the desired compound was detected. The mixture was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 2). The combined organic phases were washed with brine (5 ml x 1), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1/1) to give benzyl 4- [ [1- (1-tert-butoxycarbonylazetidine-3-carbonyl) -4-fluoro-4-piperidinyl ] methyl ] piperazine-1-carboxylate (380 mg,732.71umol,64.67% yield) as a dark brown liquid.
Step 2
To a solution of benzyl 4- [ [1- (1-tert-butoxycarbonylazetidine-3-carbonyl) -4-fluoro-4-piperidinyl ] methyl ] piperazine-1-carboxylate (380 mg,732.71umol,1 eq.) in EA (4 mL) and EtOH (4 mL) was added Pd/C (100 mg,10% purity), and the mixture was stirred at 25 ℃ under H2 (15 psi) for 12 hours. TLC (petroleum ether: ethyl acetate=0/1) showed the reaction was complete. The mixture was filtered and concentrated in vacuo. The crude product was used in the next step without further purification. The product 3- [ 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carbonyl ] azetidine-1-carboxylic acid tert-butyl ester (240 mg,624.21umol,85.19% yield) was a brown liquid.
Step 3
To a mixture of 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (100 mg,232.02umol,1 eq) and 3- [ 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carbonyl ] azetidine-1-carboxylic acid tert-butyl ester (178.42 mg,464.03umol,2 eq) in DMSO (2 mL) was added DIEA (89.96 mg,696.05umol,121.24ul,3 eq) at 25 ℃ in one portion. The mixture was stirred at 100℃for 1 hour. TLC (petroleum ether: ethyl acetate=0/1) showed the reaction was complete. The mixture was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=0/1) to give 3- [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carbonyl ] azetidine-1-carboxylic acid tert-butyl ester (180 mg,221.81umol,95.60% yield, 96% purity) as a brown oil.
Step 4
To a mixture of tert-butyl 3- [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carbonyl ] azetidine-1-carboxylate (90 mg,115.53umol,1 eq) in DCM (2 mL) was added TFA (1.54 g,13.51mmol,1mL,116.91 eq) at 25 ℃. The mixture was stirred at 25℃for 1 hour. TLC (petroleum ether: ethyl acetate=0/1) showed the reaction was complete. The mixture was concentrated at 40 ℃ under reduced pressure. The crude product was used in the next step without further purification. The product azetidin-3-yl- [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] methanone (52 mg,78.67umol,68.09% yield, 83% purity) was as a grey solid.
Step 5
To a mixture of azetidin-3-yl- [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] methanone (52 mg,94.78 mol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (31.42 mg,113.73 mol,1.2 eq.) in DMSO (2 mL) was added DIEA (36.75 mg,284.33 mol,49.53ul,3 eq.) at 25 ℃ in one portion. The mixture was stirred at 100℃for 12 hours. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃ and poured into water (15 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -45%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [3- [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carbonyl ] azetidin-1-yl ] isoindoline-1, 3-dione (10.2 mg,12.14umol,12.81% yield, 95.8% purity) as a yellow solid.
Exemplary Synthesis of Compound 137
Step 1
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-isoindoline-1, 3-dione (500 mg,1.81mmol,1 eq.) and 4- (dimethoxymethyl) piperidine (1 g,6.28mmol,3.47 eq.) in DMSO (10 mL) was added DIEA (701.79 mg,5.43mmol,945.81uL,3 eq.). The mixture was stirred at 120℃for 2 hours. The residue was diluted with H2O (50 mL) and extracted with ethyl acetate (40 mL. Times.3). The combined organic layers were washed with brine (3×30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was then dissolved in ethyl acetate (8 mL), followed by addition of petroleum ether (50 mL), followed by stirring at 25 ℃ for 16h. The mixture was filtered and the filter cake was dried and concentrated under reduced pressure to give 4- [4- (dimethoxymethyl) -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (800 mg, crude) as a yellow solid.
Step 2
A solution of 4- [4- (dimethoxymethyl) -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (800 mg,1.93mmol,1 eq.) in THF (4 mL) and HCl (2M, 4.21mL,4.37 eq.) is stirred at 70℃for 1 hour. After cooling, the reaction was diluted with water (20 mL) and neutralized to pH 7 with saturated NaHCO 3. The mixture was then filtered and the filter cake dried under reduced pressure to give 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] piperidine-4-carbaldehyde (600 mg,1.53mmol,79.29% yield, 94% purity) as a yellow solid.
Step 3
To a solution of 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (45 mg,97.49umol,1 eq.) in DCM (5 mL) was added DIEA (63.00 mg,487.43umol,84.90ul,5 eq.) and stirred at 20 ℃ for 10min. The mixture was then concentrated. The residue was stirred with a solution of 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] piperidine-4-carbaldehyde (43.21 mg,116.98umol,1.2 eq.) in HOAC (1 mL) and MeOH (10 mL) at 20 ℃ for 20min, followed by the addition of borane; 2-methylpyridine (20.85 mg,194.97umol,2 eq). The mixture was then stirred at 25 ℃ under N2 for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,35 min) to give 2- (2, 6-dioxo-3-piperidinyl) -4- [4- [ [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (33.4 mg,40.82umol,41.87% yield, 99.60% purity) as a yellow solid.
Exemplary Synthesis of Compound 138
Step 1
To a solution of 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methylpiperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (80 mg,219.51umol,1 eq) and tert-butyl 4- (2-oxoethyl) piperidine-1-carboxylate (64.86 mg,285.37umol,1.3 eq) in MeOH (10 mL) and HOAC (1 mL) was added borane; 2-methylpyridine (46.96 mg,439.03umol,2 eq.). The mixture was stirred at 25℃for 4 hours. TLC (dichloromethane: methanol=10:1, rf=0.4) showed no starting material and a new spot. The residue was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% to 10% dichloromethane/methanol) to give 4- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] piperidine-1-carboxylic acid tert-butyl ester (60 mg,92.75umol,42.25% yield, 89% purity) as a yellow solid.
Step 2
To a solution of 4- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] piperidine-1-carboxylic acid tert-butyl ester (60 mg,104.21umol,1 eq.) in DCM (3 mL) was added TFA (3.08 g,27.01mmol,2mL,259.20 eq.). The mixture was stirred at 25℃for 1 hour. TLC (dichloromethane: methanol=10:1, rf=0.01) showed no starting material and a new spot. The residue was concentrated under reduced pressure to give 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- [2- (4-piperidinyl) ethyl ] piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (60 mg, crude TFA) as a yellow gum.
Step 3
To a solution of 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- [2- (4-piperidinyl) ethyl ] piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (60 mg,101.76umol,1 eq, TFA) in DCM (5 mL) was added DIEA (65.76 mg,508.78umol,88.62ul,5 eq) and stirred at 20 ℃ for 10min. The mixture was then concentrated. The residue was stirred with a solution of 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] piperidine-4-carbaldehyde (45.10 mg,122.11umol,1.2 eq.) in HOAC (1 mL) and MeOH (10 mL) at 20 ℃ for 20min, followed by addition of borane; 2-methylpyridine (21.77 mg,203.51umol,2 eq). The mixture was then stirred at 25 ℃ under N2 for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,35 min) to give 2- (2, 6-dioxo-3-piperidinyl) -4- [4- [ [4- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (34.3 mg,40.96umol,40.25% yield, 98.99% purity) as a yellow solid.
Exemplary Synthesis of Compound 139
Step 1
To a mixture of 3-hydroxypropyl 4-methylbenzenesulfonate (500 mg,2.17mmol,1 eq) and 2- (2, 6-dioxo-3-piperidinyl) -4-hydroxy-isoindoline-1, 3-dione (595.42 mg,2.17mmol,1 eq) in DMSO (5 mL) was added Na2CO3 (690.39 mg,6.51mmol,3 eq) in one portion at 25 ℃. The mixture was stirred at 60℃for 2 hours. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃, filtered and poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (30 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,35 min) to give 2- (2, 6-dioxo-3-piperidyl) -4- (3-hydroxypropoxy) isoindoline-1, 3-dione (110 mg,331.02umol,15.25% yield, 100% purity) as a gray solid.
Step 2
To a mixture of 2- (2, 6-dioxo-3-piperidyl) -4- (3-hydroxypropoxy) isoindoline-1, 3-dione (110 mg,331.02umol,1 eq.) DMAP (8.09 mg,66.20umol,0.2 eq.) and TEA (167.48 mg,1.66mmol,230.37uL,5 eq.) in DMF (2 mL) was added 4-methylbenzenesulfonyl chloride (189.32 mg,993.06umol,3 eq.) at 0deg.C. The mixture was stirred at 25℃for 12 hours. TLC (petroleum ether: ethyl acetate=1/1) showed the reaction was complete. The mixture was poured into water (10 mL) and extracted with ethyl acetate (15 mL x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1/1) to give 3- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] oxypropyl 4-methylbenzenesulfonate (80 mg,88.80umol,26.83% yield, 54% purity) as a pale yellow oil.
Step 3
To a mixture of 3- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] oxypropyl 4-methylbenzenesulfonate (40 mg,82.22 uL, 1 eq.) and 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (56.93 mg,123.33 uL, 1.5 eq.) in MeCN (1 mL) was added DIEA (53.13 mg,411.11 uL, 71.61uL,5 eq.) and KI (13.65 mg,82.22 uL, 1 eq.) at 25 ℃. The mixture was stirred at 66 ℃ for 12 hours. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -4- [3- [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] propoxy ] isoindoline-1, 3-dione (10.5 mg,12.28umol,14.93% yield, 96.1% purity, FA) as a gray solid.
Exemplary Synthesis of Compound 140
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-isoindoline-1, 3-dione (500 mg,1.81mmol,1 eq.) and 3-aminopropan-1-ol (271.90 mg,3.62mmol,279.16ul,2 eq.) in DMSO (5 mL) was added DIEA (701.77 mg,5.43mmol,945.79ul,3 eq.) at 20℃in one portion. The mixture was stirred at 100℃for 2h. LCMS showed the reaction was complete. The mixture was cooled to 20 ℃. The aqueous phase was extracted with ethyl acetate (3X 10 mL). The combined organic phases were washed with brine (2×10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% MeOH/DCM) to give 2- (2, 6-dioxo-3-piperidinyl) -4- (3-hydroxypropylamino) isoindoline-1, 3-dione (146 mg,440.66umol,24.35% yield) as a yellow gum.
Step 2
To a mixture of 2- (2, 6-dioxo-3-piperidyl) -4- (3-hydroxypropylamino) isoindoline-1, 3-dione (126 mg,380.29umol,1 eq.) and TosCl (72.50 mg,380.29umol,1 eq.) in DCM (10 mL) was added DMAP (46.46 mg,380.29umol,1 eq.) and Et3N (76.96 mg,760.59umol,105.86ul,2 eq.) at 25 ℃ in N2. The mixture was stirred at 25℃for 2 hours. TLC showed the reaction was complete. The residue was poured into water (10 mL). The aqueous phase was extracted with DCM (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -100% (20 min) ethyl acetate/petroleum ether) to give 4-methylbenzenesulfonic acid 3- [ [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-4-yl ] amino ] propyl ester (98 mg,189.74umol,49.89% yield, 94% purity) as a yellow gum.
Step 3
To a mixture of 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -1H-indazole (60 mg,129.98umol,1 eq.) and 4-methylbenzenesulfonic acid 3- [ [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] amino ] propyl ester (98 mg,201.85umol,1.55 eq.) in MeCN (5 mL) were added DIPEA (84.00 mg,649.91umol,113.20ul,5 eq.) and KI (107.89 mg,649.91umol,5 eq.) at 20 ℃. The mixture was stirred at 80℃for 16h. LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; beginning B:0; ending B:35; flow rate: 35mL/min; gradient time: 35min;100% B hold time: 1 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -4- [3- [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] propylamino ] isoindoline-1, 3-dione (50.1 mg,64.11umol,49.32% yield, 99.16% purity) as a yellow solid.
Exemplary Synthesis of Compound 141
Step 1
To a solution of benzyl 4- (4-piperidylmethyl) piperazine-1-carboxylate (1.9 g,4.40mmol,1 eq., TFA) and 4- (tert-butoxycarbonylamino) butyl 4-methylbenzenesulfonate (1.6 g,4.66mmol,1.06 eq.) in CH3CN (15 mL) was added KI (1.46 g,8.81mmol,2 eq.) and DIEA (2.85 g,22.02mmol,3.84mL,5 eq.). After the addition, the reaction mixture was stirred at 80 ℃ for 12h. LCMS showed the desired MS. TLC (dichloromethane: methanol=10:1) showed several new spots. After cooling, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (3×20 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% methanol/dichloromethane) to give benzyl 4- [ [1- [4- (tert-butoxycarbonylamino) butyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (680 mg,1.15mmol,26.10% yield, 82.6% purity) as a yellow oil.
Step 2
To a solution of benzyl 4- [ [1- [4- (tert-butoxycarbonylamino) butyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (680 mg,1.39mmol,1 eq.) in DCM (5 mL) was added TFA (4.62 g,40.52mmol,3mL,29.12 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 1h. LCMS showed starting material was consumed and the desired MS was found. The reaction mixture was concentrated under reduced pressure to give benzyl 4- [ [1- (4-aminobutyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (840 mg, crude, TFA) as a yellow oil. The crude product was used directly in the next step.
Step 3
To a solution of benzyl 4- [ [1- (4-aminobutyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (840 mg,1.67mmol,1 eq., TFA) in DMF (10 mL) was added 2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-isoindoline-1, 3-dione (461.68 mg,1.67mmol,1 eq.) and DIEA (1.08 g,8.36mmol,1.46mL,5 eq.). After the addition, the reaction mixture was stirred at 90 ℃ for 12h. LCMS showed the desired MS. TLC (dichloromethane: methanol=10:1) showed several new spots. After addition, the reaction was diluted with ethyl acetate (20 mL) and washed with brine (2×20 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% methanol/dichloromethane) to give benzyl 4- [ [1- [4- [ [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] amino ] butyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (84 mg,111.72umol,6.68% yield, 85.75% purity) as a yellow solid.
Step 4
A mixture of 4- [ [1- [4- [ [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] amino ] butyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (84 mg,130.28umol,1 eq.) and TFA (2 mL) was stirred at 80℃for 1h. TLC (dichloromethane: methanol=5:1) showed that the starting material was consumed and a new spot formed. The reaction mixture was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -4- [4- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] butylamino ] isoindoline-1, 3-dione (81 mg, crude, TFA) as a yellow gum. The crude product was used directly in the next step.
Step 5
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -4- [4- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] butylamino ] isoindoline-1, 3-dione (81.00 mg,129.67umol,1 eq., TFA) and 3- (6-chloropyrimidin-4-yl) -6- (1-methylcyclopropoxy) -1H-indazole (45 mg,149.63umol,1.15 eq.) in DMSO (2 mL) was added DIEA (83.80 mg,648.36umol,112.93ul,5 eq.). After addition, the reaction was stirred at 90 ℃ for 24h to give a brown solution. LCMS showed starting material was consumed and the desired MS was found. The reaction mixture was diluted with water (15 mL) and extracted with ethyl acetate (3×10 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -30%;40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -4- [4- [4- [ [4- [6- [6- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] butylamino ] isoindoline-1, 3-dione (13.8 mg,17.32umol,13.36% yield, 97.27% purity) as a yellow solid.
Exemplary Synthesis of Compound 142
Step 1
To a mixture of 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (200 mg,217.94umol,1 eq., 4 TFA) and 3-bromo-1, 1-dimethoxy-propane (79.78 mg,435.88umol,59.54ul,2 eq.) in DMF (5 mL) was added DIEA (225.34 mg,1.74mmol,303.69ul,8 eq.) at 25 ℃. The mixture was stirred at 90℃for 2 hours. TLC (dichloromethane: methanol=10/1) showed the reaction was complete. The mixture was cooled to 25 ℃. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 2). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10/1) to give 3- [6- [ (3S) -4- [ [1- (3, 3-dimethoxypropyl) -4-piperidinyl ] methyl ] -3-methyl-piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1H-indazole (70 mg,120.45umol,55.27% yield, 97% purity) as a pale yellow oil.
Step 2
To a mixture of 3- [6- [ (3S) -4- [ [1- (3, 3-dimethoxypropyl) -4-piperidinyl ] methyl ] -3-methyl-piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1H-indazole (70 mg,124.17umol,1 eq.) in THF (1 mL) was added H2SO4 (2 m,310.43ul,5 eq.). The mixture was stirred at 60℃for 1 hour. LCMS showed the reaction was complete. The mixture was poured into water (1 mL). The pH of the mixture was adjusted to 7-8 by adding Na2CO 3. The mixture was extracted with DCM (5 ml x 3). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated in vacuo to give 3- [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] propanal as a brown oil (56 mg,106.01umol,85.38% yield, 98% purity). The crude product was used in the next step without further purification.
Step 3
To a mixture of 3- (4-amino-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (42.07 mg,162.27umol,1.5 eq.) in HOAc (0.3 mL) and MeOH (3 mL) was added 3- [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] propanal (56 mg,108.18umol,1 eq.) at 25 ℃ in one portion followed by borane; 2-methylpyridine (23.14 mg,216.36umol,2 eq). The mixture was stirred at 25℃for 12 hours. LCMS showed 44% of the desired product. The mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (column: phenomenex Luna C, 75X 30mm X3 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,35 min) to give 3- [4- [3- [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] propylamino ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (13.2 mg,16.19umol,14.97% yield, 99% purity, FA) as a gray solid.
Exemplary Synthesis of Compound 143
Step 1
To a solution of benzyl 4- (4-piperidinylmethyl) piperazine-1-carboxylate (3 g,8.48mmol,1 eq, HCl) and 4-bromobutan-1-ol (1.62 g,8.48mmol,80% purity, 1 eq) in THF (10 mL) was added K2CO3 (3.51 g,25.43mmol,3 eq). The mixture was then stirred at 60 ℃ under N2 for 16 hours. LCMS showed no starting material. TLC (dichloromethane: methanol=10:1, rf=0.2) showed a new spot for the reaction. The reaction mixture was filtered and the filtrate was concentrated to give benzyl 4- [ [1- (4-hydroxybutyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (3.1 g, crude) as a white gum.
Step 2
A solution of oxalyl chloride (716.88 mg,5.65mmol,494.40uL,1.1 eq.) in DCM (10 mL) was cooled to-60℃under a dry nitrogen atmosphere. A solution of DMSO (1.00 g,12.84mmol,1.00mL,2.5 eq.) in DCM (10 mL) was added dropwise, and the mixture was subsequently stirred at-60℃for 15min. Next, a solution of benzyl 4- [ [1- (4-hydroxybutyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (2 g,5.13mmol,1 eq.) in DCM (10 mL) was added dropwise and the mixture stirred at-60℃for 45min. Subsequently, TEA (1.56 g,15.40mmol,2.14mL,3 eq.) was added and the mixture was warmed to-60℃for 1h. TLC (dichloromethane: methanol=10:1, rf=0.4) showed a new spot for the reaction. The reaction mixture was filtered and the filtrate was used directly in the next step, benzyl 4- [ [1- (4-oxobutyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (1.5 g, crude) was used directly in the next step as a yellow liquid in DCM solution.
Step 3
To a solution of benzyl 4- [ [1- (4-oxobutyl) -4-piperidinyl ] methyl ] piperazine-1-carboxylate (1.5 g,3.87mmol,1.43 eq.) and 3- (4-amino-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (700 mg,2.70mmol,1 eq.) in DCM (15 mL) and MeOH (15 mL) was added HOAc (16.21 mg,270.00umol,15.44ul,0.1 eq.) and the mixture stirred at 20 ℃ for 20min. To this solution was then added NaBH3CN (509.00 mg,8.10mmol,3 eq.) and stirred at 20 ℃ for 16 hours. TLC (dichloromethane: methanol=10:1, rf=0.2) showed the reaction was complete. The reaction mixture was poured into H2O (20 mL). The mixture was extracted with ethyl acetate (30 ml x 3). The organic phase was washed with brine (20 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (0% -20% methanol/dichloromethane) to give the compound. The residue was purified by preparative HPLC (column: xtime C18:40 mm 10um; mobile phase: [ water (0.225% FA) -ACN ]; B%:5% -35%,10 min) to give benzyl 4- [1- [4- [ [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-4-yl ] amino ] butyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (300 mg,461.34umol,17.09% yield, 97% purity) as a white solid.
Step 4
To a solution of benzyl 4- [ [1- [4- [ [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-4-yl ] amino ] butyl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (100 mg,158.54umol,1 eq.) was added TFA (4.62 g,40.52mmol,3ml,255.58 eq.) and the mixture was stirred at 70 ℃ for 1h. TLC (dichloromethane: methanol=10:1, rf=0.01) showed the reaction was complete. The reaction mixture was concentrated in vacuo to give 3- [ 1-oxo-4- [4- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] butylamino ] isoindolin-2-yl ] piperidine-2, 6-dione (78 mg, crude) as a colourless gum.
Step 5
To a solution of 3- (6-chloropyrimidin-4-yl) -6- (1-methylcyclopropoxy) -1H-indazole (35 mg,116.38umol,7.41e-1 eq.) and 3- [ 1-oxo-4- [4- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] butylamino ] isoindol-2-yl ] piperidine-2, 6-dione (78 mg,157.05umol,1 eq.) in DMSO (5 mL) and DIEA (202.98 mg,1.57mmol,273.55uL,10 eq.). The mixture was stirred at 80℃for 16h. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -30%,40 min) to give 3- [4- [4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] butylamino ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (8.3 mg,10.91umol,6.95% yield, 100% purity) as a white solid.
Exemplary Synthesis of Compound 144
Step 1
To a solution of tert-butyl 3- (aminomethyl) -3-hydroxy-azetidine-1-carboxylate (5 g,24.72mmol,1 eq.) in DCM (50 mL) was added Et3N (3.75 g,37.08mmol,5.16mL,1.5 eq.) followed by dropwise addition of 2-chloroacetyl chloride (3.35 g,29.67mmol,2.36mL,1.2 eq.) at 0deg.C. The mixture was stirred at 25℃for 2 hours. LCMS showed the desired MS. TLC (dichloromethane: methanol=10:1) showed several spots. The reaction mixture was diluted with water (40 mL) and extracted with dichloromethane (2×50 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 5% methanol/dichloromethane) to give 3- [ [ (2-chloroacetyl) amino ] methyl ] -3-hydroxy-azetidine-1-carboxylic acid tert-butyl ester (4 g,14.35mmol,58.05% yield, 100% purity) as a white solid.
Step 2
To a solution of tert-butyl 3- [ [ (2-chloroacetyl) amino ] methyl ] -3-hydroxy-azetidine-1-carboxylate (3 g,10.76mmol,1 eq.) in dioxane (30 mL) was added NaH (753.34 mg,18.84mmol,60% purity, 1.75 eq.) at 25 ℃. The reaction solution was stirred at 80℃for 12h. TLC (dichloromethane: methanol=10:1) showed several new spots. After cooling, the reaction mixture was quenched with water (20 mL) and extracted with ethyl acetate (15 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% methanol/dichloromethane) to give 7-oxo-5-oxa-2, 8-diazaspiro [3.5] nonane-2-carboxylic acid tert-butyl ester (520 mg,2.15mmol,19.94% yield) as a white solid.
Step 3
To a solution of 7-oxo-5-oxa-2, 8-diazaspiro [3.5] nonane-2-carboxylic acid tert-butyl ester (520 mg,2.15mmol,1 eq.) in THF (8 mL) was added bh3.THF (1 m,6.44mL,3 eq.) at 0 ℃ under N2. After the addition, the reaction solution was stirred at 25 ℃ for 12h. TLC (dichloromethane: methanol=10:1) showed several new spots. The solution was quenched by dropwise addition of 15% sodium hydroxide solution (10 mL) over a period of 5 minutes to control gas evolution. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (20 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% methanol/dichloromethane) to give tert-butyl 5-oxa-2, 8-diazaspiro [3.5] nonane-2-carboxylate (430 mg,1.88mmol,87.76% yield) as a white solid.
Step 4
To a solution of 4-bromo-2-fluoropyridine (331.49 mg,1.88mmol,1 eq.) in DMSO (5 mL) was added tert-butyl 5-oxa-2, 8-diazaspiro [3.5] nonane-2-carboxylate (430 mg,1.88mmol,1 eq.) and K2CO3 (520.65 mg,3.77mmol,2 eq.). After the addition, the reaction solution was stirred at 90 ℃ for 12h. TLC (petroleum ether: ethyl acetate=3:1) showed several new spots. After cooling, the reaction mixture was filtered, and the filtrate was diluted with ethyl acetate (50 mL) and washed with brine (3×30 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether) to give tert-butyl 8- (4-bromo-2-pyridinyl) -5-oxa-2, 8-diazaspiro [3.5] nonane-2-carboxylate (430 mg,1.05mmol,55.84% yield, 94% purity) as a colorless oil.
Step 5
To a solution of tert-butyl 8- (4-bromo-2-pyridinyl) -5-oxa-2, 8-diazaspiro [3.5] nonane-2-carboxylate (560 mg,1.46mmol,1 eq.) in dioxane (10 mL) was added Pin2B2 (740.14 mg,2.91mmol,2 eq.), KOAc (429.06 mg,4.37mmol,3 eq.) and Pd (dppf) Cl2 (106.63 mg,145.73umol,0.1 eq.). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 12h. LCMS showed the starting material was consumed and the desired MS formed. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give [2- (2-tert-butoxycarbonyl-5-oxa-2, 8-diazaspiro [3.5] non-8-yl) -4-pyridinyl ] boronic acid (800 mg, crude product) as a black oil. The crude product was used directly in the next step.
Step 6
To a solution of [2- (2-tert-butoxycarbonyl-5-oxa-2, 8-diazaspiro [3.5] non-8-yl) -4-pyridinyl ] boronic acid (800 mg,2.29mmol,1.85 eq.) in dioxane (10 mL) and H2O (2 mL) was added 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (550 mg,1.24mmol,1 eq.), na2CO3 (393.54 mg,3.71mmol,3 eq.) and Pd (dppf) Cl2 (90.56 mg,123.77 mol,0.1 eq.). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 4h. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=5:1) showed several new spots. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether) to give 8- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -5-oxa-2, 8-diazaspiro [3.5] nonane-2-carboxylic acid tert-butyl ester (554 mg,825.55umol,66.70% yield, 92% purity) as a yellow gum.
Step 7
To a solution of 8- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -5-oxa-2, 8-diazaspiro [3.5] nonane-2-carboxylic acid tert-butyl ester (5538 mg,897.34umol,1 eq.) in DCM (3 mL) was added TFA (4.62 g,40.52mmol,3mL,45.15 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 12h. LCMS showed starting material was consumed. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in dioxane (10 mL) and ammonium hydroxide (5 mL) was added. The mixture was then stirred at 25℃for 30min. The mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure to give 8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -5-oxa-2, 8-diazaspiro [3.5] nonane (220 mg,485.56umol,54.11% yield, 86.4% purity) as a yellow solid.
Step 8
To a solution of 8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -5-oxa-2, 8-diazaspiro [3.5] nonane (220 mg,561.99umol,1 eq.) in MeOH (5 mL) were added 4-formylpiperidine-1-carboxylic acid tert-butyl ester (239.72 mg,1.12mmol,2 eq.) and borane; 2-methylpyridine (120.22 mg,1.12mmol,2 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 12h. LCMS showed the starting material was consumed and the desired MS formed. TLC (dichloromethane: methanol=10:1) showed several new spots. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% methanol/dichloromethane) to give 4- [ [8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -5-oxa-2, 8-diazaspiro [3.5] non-2-yl ] methyl ] piperidine-1-carboxylic acid tert-butyl ester (200 mg,328.50umol,58.45% yield, 96.7% purity) as a pale yellow solid.
Step 9
To a solution of tert-butyl 4- [ [8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -5-oxa-2, 8-diazaspiro [3.5] non-2-yl ] methyl ] piperidine-1-carboxylate (100 mg,169.85umol,1 eq) in DCM (2 mL) was added TFA (1.54 g,13.51mmol,1mL,79.52 eq). After the addition, the reaction solution was stirred at 25 ℃ for 1h. TLC (dichloromethane: methanol=10:1) showed that the starting material was consumed and a new spot formed. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in DCM (5 mL) and treated with DIEA (1.5 mL). The mixture was concentrated under reduced pressure to give 8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -2- (4-piperidinylmethyl) -5-oxa-2, 8-diazaspiro [3.5] nonane (90 mg, crude) as a red solid. The crude product was used directly in the next step.
Step 10
To a solution of 8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -2- (4-piperidinylmethyl) -5-oxa-2, 8-diazaspiro [3.5] nonane (45 mg,92.10umol,1 eq) in MeOH (5 mL) and HOAc (0.5 mL) was added 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-4-yl ] piperidine-4-carbaldehyde (34.02 mg,92.10umol,1 eq) and borane; 2-methylpyridine (19.70 mg,184.20umol,2 eq). After the addition, the reaction solution was stirred at 25 ℃ for 12h. LCMS showed the starting material was consumed and the desired MS formed. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%;40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -4- [4- [ [4- [ [8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -5-oxa-2, 8-diazaspiro [3.5] non-2-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (29.4 mg,34.84umol,37.82% yield, 99.77% purity) as a yellow solid.
Exemplary Synthesis of Compound 145
Step 1
To a solution of methyl 3-bromo-2- (bromomethyl) benzoate (1 g,3.25mmol,1 eq) and 3-aminopiperidine-2, 6-dione (641.33 mg,3.90mmol,1.2 eq, HCl) in MeCN (10 mL) was added DIEA (2.10 g,16.24mmol,2.83mL,5 eq) and stirred at 90 ℃ for 16h. LCMS showed all desired products. The resulting solid was collected by filtration. The solid was washed with MeCN (5 ml×3) and then concentrated under reduced pressure to give 3- (4-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (900 mg, crude) as an off-white solid.
Step 2
To a solution of 3- (4-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (300 mg,928.39umol,1 eq.) and 4- (dimethoxymethyl) piperidine (443.47 mg,2.79mmol,3 eq.) in DMSO (5 mL) was added Pd-PEPSI-pentCl-O-methylpyridine (50.41 mg,92.84umol,0.1 eq.) and Cs2CO3 (604.97 mg,1.86mmol,2 eq.) and stirred at 80℃under N2 for 16h. TLC (petroleum ether: ethyl acetate=0:1, rf=0.3) showed a new spot for the reaction. The reaction was quenched with NH4Cl (20 mL) and extracted with ethyl acetate (3 x 20 mL). The combined organic phases were washed with water, dried over Na2SO4 and concentrated in vacuo to give a residue. The residue was taken up by preparative TLC (ethyl acetate. Rf=0.3) to give 3- [4- [4- (dimethoxymethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (172 mg,304.19umol,32.77% yield, 71% purity) as a white solid.
Step 3
A solution of 3- [4- [4- (dimethoxymethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (100 mg,249.09umol,1 eq.) in THF (2 mL) and HCl (2M, 6.67mL,53.53 eq.) was stirred at 20℃for 8 hours. The reaction mixture was poured into H2O (20 mL) and basified with aqueous NaHCO3 to ph=8. The mixture was extracted with ethyl acetate (20 ml x 5) and dried over anhydrous Na2SO4 and concentrated in vacuo to give 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-4-yl ] piperidine-4-carbaldehyde (70 mg, crude) as a yellow solid.
Step 4
A mixture of 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-4-yl ] piperidine-4-carbaldehyde (70 mg,98.48 mol,50% purity, 1 eq.) and 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -2H-indazole (45.46 mg,98.48 mol,1 eq.) in HOAC (1 mL) and MeOH (10 mL) was stirred at 25℃for 20min, followed by the addition of borane; 2-methylpyridine (21.07 mg,196.96umol,2 eq). The mixture was then stirred at 25 ℃ under N2 for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (10 mM NH4HCO 3) -ACN ]; B%:30% -90%,40 min) to give 3- [4- [4- [ [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (14.8 mg,18.48umol,18.76% yield, 100% purity) as a white solid.
Exemplary Synthesis of Compound 146
Step 1
To a mixture of 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -2H-indazole (48 mg,107.24umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] piperidine-4-carbaldehyde (51.50 mg,139.42umol,1.3 eq) in MeOH (5 mL) at 25 ℃ was added AcOH (1 mL) and borane in one portion; 2-methylpyridine (22.94 mg,214.49umol,2 eq). The mixture was stirred at 25℃for 4h. LCMS showed the desired MS. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow rate: 25mL/min; gradient time: 35min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -4- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (42 mg,52.44umol,48.90% yield, 100% purity) as a yellow solid.
Exemplary Synthesis of Compound 147
Step 1
A mixture of 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -2H-indazole (31.49 mg,70.35 mol,0.5 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-4-yl ] piperidine-4-carbaldehyde (50 mg,140.69 mol,1 eq.) in HOAC (1 mL) and MeOH (10 mL) was stirred at 25℃for 20min, followed by the addition of borane; 2-methylpyridine (30.10 mg,281.38umol,2 eq). The mixture was then stirred at 25 ℃ under N2 for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -35%,40 min) to give 3- [4- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (35 mg,43.83umol,31.16% yield, 98.56% purity) as a white solid.
Exemplary Synthesis of Compound 148
Step 1
To a mixture of benzyl 4- [2, 2-difluoro-2- (4-piperidinyl) ethyl ] piperazine-1-carboxylate (117.88 mg,320.82umol,1 eq), 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] piperidine-4-carbaldehyde (71.10 mg,192.49umol,0.6 eq) in MeOH (10 mL) was added AcOH (1 mL), borane; 2-methylpyridine (34.32 mg, 320.82. Mu. Mol,1 eq.) the mixture was then stirred at 25℃for 12h. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=3:1) showed complete consumption of starting material and found two new principal points. The reaction mixture was concentrated under reduced pressure to give a residue. The crude material was purified by flash chromatography on silica gel (0% -50% ethyl acetate/petroleum ether) to give 4- [2- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] -4-piperidinyl ] methyl ] -4-piperidinyl ] -2, 2-difluoro-ethyl ] piperazine-1-carboxylic acid benzyl ester (115 mg, crude) as a yellow solid.
Step 2
To a mixture of 4- [2- [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-4-yl ] -4-piperidinyl ] methyl ] -4-piperidinyl ] -2, 2-difluoro-ethyl ] piperazine-1-carboxylic acid benzyl ester (115 mg,159.54umol,1 eq) was added TFA (18.19 mg,159.54umol,11.81ul,1 eq), and the mixture was stirred at 70 ℃ for 1h. TLC (petroleum ether: ethyl acetate=3:1) showed complete consumption of starting material and found a new main point. The reaction mixture was concentrated in vacuo to give 4- [4- [ [4- (1, 1-difluoro-2-piperazin-1-yl-ethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (110 mg, crude TFA) as a brown oil.
Step 3
To a solution of 4- [4- [ [4- (1, 1-difluoro-2-piperazin-1-yl-ethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (93 mg,158.52umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (47.67 mg,158.52umol,1 eq.) in DMSO (10 mL) was added DIEA (102.44 mg,792.61umol,138.05ul,5 eq.). The mixture was stirred at 70 ℃ under N2 for 16 hours and cooled. The residue was concentrated under reduced pressure to give a residue. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to afford 4- [4- [ [4- [1, 1-difluoro-2- [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] -1-piperidinyl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (15.5 mg,17.85umol,11.26% yield, 98% purity) as a yellow solid.
Exemplary Synthesis of Compound 149
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (230 mg,832.67umol,9.47e-1 eq.) and tert-butyl 4- (4-piperidyloxy) piperidine-1-carboxylate (250 mg,879.06umol,1 eq.) in DMSO (5 mL) was added DIEA (340.83 mg,2.64mmol,459.34uL,3 eq.) at 20℃in one portion. The mixture was stirred at 100℃for 16h. LCMS showed the reaction was complete. The mixture was cooled to 20 ℃. The residue was poured into HCl (2M) to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (3X 10 mL). The combined organic phases were washed with brine (2×10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% MeOH/DCM) to give tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] piperidine-1-carboxylate (420 mg,776.91umol,88.38% yield) as a yellow gum.
Step 2
To a mixture of tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] piperidine-1-carboxylate (440.58 mg,814.97umol,1 eq.) in DCM (5 mL) at 25℃was added TFA (3.08 g,27.01mmol,2mL,33.15 eq.) in one portion. The mixture was stirred at 25 ℃ for 2 hours to give a yellow solution. TLC showed the reaction was complete. The residue was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (4-piperidinyloxy) -1-piperidinyl ] isoindoline-1, 3-dione (500 mg,732.94umol,89.94% yield, 98% purity, 2 TFA) as a yellow oil.
Step 3
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (4-piperidinyloxy) -1-piperidinyl ] isoindoline-1, 3-dione (180 mg,408.63umol,1 eq.) and (2R) -tert-butyl 2- (p-toluenesulfonyloxymethyl) morpholine-4-carboxylate (227.68 mg,612.95umol,1.5 eq.) in MeCN (10 mL) was added KI (339.17 mg,2.04mmol,5 eq.) and DIPEA (264.07 mg,2.04mmol,355.88uL,5 eq.) at 20℃in one portion. The mixture was stirred at 100 ℃ for 16 hours to give a yellow suspension. LCMS showed the desired MS. The mixture was cooled to 25 ℃ and concentrated under reduced pressure at 25 ℃. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (3X 20 mL). The combined organic phases were washed with brine (2×20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (column: 12g,100-200 mesh silica gel, 0% -10% (5 min) MeOH/DCM,10% (10 min) MeOH/DCM) to give tert-butyl (2S) -2- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] oxy ] -1-piperidinyl ] methyl ] morpholine-4-carboxylate (255 mg,362.73umol,88.77% yield, 91% purity) as a yellow gum.
Step 4
To a solution of tert-butyl (2S) -2- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] -1-piperidinyl ] methyl ] morpholine-4-carboxylate (255 mg,398.60umol,1 eq.) in DCM (5 mL) was added TFA (3.08 g,27.01mmol,2mL,67.77 eq.) at 25 ℃ in one portion. The mixture was stirred at 25℃for 20min to give a yellow solution. LCMS (EB 16-910-P1 A1) showed that the reaction was complete. The solution was concentrated in vacuo to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [ [ (2R) -morpholin-2-yl ] methyl ] -4-piperidinyl ] oxy ] -1-piperidinyl ] isoindoline-1, 3-dione (330 mg,344.34umol,86.39% yield, 92% purity, 3 TFA) as a yellow gum.
Step 5
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [ [ (2R) -morpholin-2-yl ] methyl ] -4-piperidinyl ] oxy ] -1-piperidinyl ] isoindoline-1, 3-dione (330 mg,374.28 mol,1.13 eq, 3 TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (100 mg,332.51 mol,1 eq) in DMSO (5 mL) was added DIEA single portion (343.79 mg,2.66mmol,463.33ul,8 eq) at 20 ℃. The mixture was stirred at 80℃for 36h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:35; flow rate: 25mL/min; gradient time: 35min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [1- [ [ (2S) -4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] methyl ] -4-piperidinyl ] oxy ] -1-piperidinyl ] isoindoline-1, 3-dione (45.5 mg,55.01umol,16.54% yield, 97.19% purity) as a yellow solid.
Exemplary Synthesis of Compound 150
Compound 150 was prepared in a similar manner to compound 149 using and (2S) -2- (p-tolylsulfonyloxymethyl) morpholine-4-carboxylic acid tert-butyl ester.
Exemplary Synthesis of Compound 151
Step 1
To a solution of 8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -2- (4-piperidinylmethyl) -5-oxa-2, 8-diazaspiro [3.5] nonane (45 mg,92.10umol,1 eq) in MeOH (5 mL) and HOAc (0.5 mL) was added 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] piperidine-4-carbaldehyde (34.02 mg,92.10umol,1 eq) and borane; 2-methylpyridine (19.70 mg, 184.19. Mu. Mol,2 eq). After the addition, the reaction solution was stirred at 25 ℃ for 12h. LCMS showed the starting material was consumed and the desired MS formed. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%;40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [8- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -5-oxa-2, 8-diazaspiro [3.5] non-2-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (37.3 mg,43.97umol,47.75% yield, 99.26% purity) as a yellow solid.
Exemplary Synthesis of Compound 152
Step 1
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindoline-1, 3-dione (300 mg, 541.98. Mu.mol, 1.34 eq., TFA) and tert-butyl (2S) -2- (p-toluenesulfonyloxymethyl) morpholine-4-carboxylate (150 mg, 403.82. Mu.mol, 1 eq.) in CH3CN (5 mL) was added DIEA (260.96 mg,2.02mmol, 351.69. Mu.L, 5 eq.) and KI (134.07 mg, 807.65. Mu.mol, 2 eq.). After the addition, the reaction solution mixture was stirred at 80 ℃ for 12h. LCMS showed the desired MS. TLC (dichloromethane: methanol=10:1) showed several new spots. After cooling, the reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (3×10 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% methanol in dichloromethane) to give tert-butyl (2R) -2- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] morpholine-4-carboxylate (107 mg,150.76umol,37.33% yield, 90% purity) as a yellow solid.
Step 2
To a solution of tert-butyl (2R) -2- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] morpholine-4-carboxylate (105 mg,164.38umol,1 eq.) in DCM (5 mL) was added TFA (4.62 g,40.52mmol,3mL,246.49 eq.). After the addition, the reaction solution mixture was stirred at 25 ℃ for 1h. LCMS showed the desired MS. The reaction mixture was concentrated under pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (2S) -morpholin-2-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (110 mg, crude, TFA) as a yellow solid. The crude product was used directly in the next step.
Step 3
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (2S) -morpholin-2-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (88 mg,134.83 mol,1.01 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (40 mg,133.00 mol,1 eq) in DMSO (1.5 mL) was added DIEA (85.95 mg,665.02 mol,115.83ul,5 eq). After the addition, the reaction solution was stirred at 90 ℃ for 16h. LCMS showed the main desired MS. After cooling, the reaction mixture was diluted with ethyl acetate (10 mL) and washed with brine (2×5 mL). The organic layer was concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%;40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (2R) -4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (22.1 mg,27.40umol,20.60% yield, 99.53% purity) as a yellow solid.
Exemplary Synthesis of Compound 153
Compound 153 was prepared in a similar manner to compound 152 using and (2R) -2- (p-tolylsulfonyloxymethyl) morpholine-4-carboxylic acid tert-butyl ester.
Exemplary Synthesis of Compound 154
Step 1
To a mixture of 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (1 g,2.32mmol,1 eq.) and 2-morpholin-2-ylethanol (304.34 mg,2.32mmol,1 eq.) in DMSO (10 mL) was added Et3N (704.34 mg,6.96mmol,968.82uL,3 eq.) in one portion followed by stirring at 100℃for 1h. TLC showed complete consumption of starting material. The mixture was cooled to 20 ℃, then the residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (3X 10 mL). The combined organic phases were washed with brine (2×10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -10% (10 min) ethyl acetate/petroleum ether, 10% (5 min) ethyl acetate/petroleum ether) to give 2- [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] ethanol as a yellow oil (1.1 g,2.09mmol,90.18% yield).
Step 2
To a mixture of 2- [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] ethanol (1.1 g,2.09mmol,1 eq.) and 4-methylbenzenesulfonyl chloride (796.90 mg,4.18mmol,2 eq.) in DCM (5 mL) at 20℃was added TEA (211.49 mg,2.09mmol,290.90uL,1 eq.) and DMAP (255.34 mg,2.09mmol,1 eq.) in one portion. The mixture was stirred at 20℃for 1h to give a yellow solution. TLC (petroleum ether: ethyl acetate=3:1, rf=0.23, ua 254 nm) showed the reaction was complete. The residue was poured into water (5 mL). The aqueous phase was extracted with DCM (5 ml x 3). The combined organic phases were washed with brine (5 ml x 3), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -10% (10 min) ethyl acetate/petroleum ether, 10% (10 min) ethyl acetate/petroleum ether) to give 4-methylbenzenesulfonic acid 2- [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] ethyl ester as a yellow solid (830 mg,1.22mmol,58.41% yield).
Step 3
To a mixture of 2- [4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] ethyl 4-methylbenzenesulfonate (830 mg,1.22mmol,1 eq.) and tert-butyl piperazine-1-carboxylate (454.74 mg,2.44mmol,2 eq.) in MeCN (10 mL) at 20 ℃ was added KI (405.29 mg,2.44mmol,2 eq.) and DIPEA (315.54 mg,2.44mmol,425.26ul,2 eq.) at once. The mixture was stirred at 80℃for 2 hours. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -40% (15 min) ethyl acetate/petroleum ether, 40% (5 min) ethyl acetate/petroleum ether) to give tert-butyl 4- [2- [4- [6- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] ethyl ] piperazine-1-carboxylate (800 mg,1.15mmol,94.43% yield) as a yellow gum.
Step 4
To a mixture of tert-butyl 4- [2- [4- [6- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] ethyl ] piperazine-1-carboxylate (800 mg,1.15mmol,1 eq.) in MeOH (5 mL) was added HCl/dioxane (4 m,5.72mL,19.84 eq.) at 25 ℃ in one portion. The mixture was stirred at 65℃for 1h. TLC showed the reaction was complete. The residue was adjusted to ph=9-10 and the aqueous phase was extracted with DCM (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -2- (2-piperazin-1-ylethyl) morpholine (500 mg,1.06mmol,91.69% yield, 98% purity) as a yellow gum.
Step 5
4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -2- (2-piperazin-1-ylethyl) morpholine (500 mg,1.08mmol,1 eq) was isolated by SFC (column DAICEL CHIRALPAK IE (250 mm. Times.30 mm,10 um); condition: 0.1% NH3H2O ETOH; start B:55; end B:55; flow: 80 mL/min) to give (2S) -4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -2- (2-piperazin-1-ylethyl) morpholine or its enantiomer (212 mg, 71 mg) (Rt=2.224 min,212 mg) and (2R) -4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -2- (2-piperazin-1-ylethyl) morpholine or its enantiomer (291 mg ) as a yellow solid.
Step 6
To a mixture of (2R) -4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -2- (2-piperazin-1-ylethyl) morpholine or its enantiomer (100.00 mg,215.72umol,1 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (79.68 mg,215.72umol,1 eq.) in MeOH (5 mL) and HOAc (0.5 mL) at 20 ℃ was added borane in one portion; 2-methylpyridine (46.15 mg,431.43umol,2 eq). The mixture was stirred at 30℃for 16h. LCMS showed the desired MS. The residue was poured into water (2 mL). The aqueous phase was extracted with ethyl acetate (2 ml x 3). The combined organic phases were washed with brine (2 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [2- [ (2S) -4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] morpholin-2-yl ] ethyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione or enantiomer thereof (59.6 mg,72.73umol,33.72% yield, 99.69% purity) as a yellow solid.
Exemplary Synthesis of Compound 155
Compound 155 is prepared in a similar manner to compound 154 using (2S) -4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -2- (2-piperazin-1-ylethyl) morpholine or an enantiomer thereof.
Exemplary Synthesis of Compound 156
Step 1
To a solution of 5- (1-methylcyclopropoxy) -3- (6-piperazin-1-ylpyrimidin-4-yl) -1H-indazole (300 mg,775.44umol,1 eq, HCl) and tert-butyl (2S) -2- (p-toluenesulfonyloxymethyl) morpholine-4-carboxylate (500 mg,1.35mmol,1.74 eq) in CH3CN (5 mL) were added DIEA (501.10 mg,3.88mmol,675.34ul,5 eq) and KI (386.17 mg,2.33mmol,3 eq). After the addition, the reaction mixture was stirred at 80 ℃ for 12h. The reaction mixture was then heated to 100 ℃ for 12h. TLC (dichloromethane: methanol=10:1) showed one major new spot. The reaction mixture was diluted with water (15 mL) and extracted with dichloromethane (3×10 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 6% methanol/dichloromethane) to give (2R) -2- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] morpholine-4-carboxylic acid tert-butyl ester (450 mg,736.82umol,95.02% yield, 90% purity) as a pale yellow solid.
Step 2
To a solution of tert-butyl (2R) -2- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] morpholine-4-carboxylate (450 mg,818.68umol,1 eq.) in DCM (3 mL) was added TFA (4.62 g,40.52mmol,3mL,49.49 eq.). After the addition, the reaction solution was stirred at 20 ℃ for 1h. LCMS showed starting material was consumed and the desired MS was found. The reaction mixture was concentrated under reduced pressure. The resulting mixture was dissolved in dichloromethane (3 mL) and treated with DIEA (1 mL). The mixture was concentrated in vacuo to give (2S) -2- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] morpholine (340 mg, crude) as a yellow solid. The crude product was used directly in the next step.
Step 3
To a solution of (2S) -2- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] morpholine (162.00 mg,360.36umol,1.48 eq) in MeOH (3 mL), DMSO (0.5 mL) and HOAc (0.5 mL) was added 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] piperidine-4-carbaldehyde (90 mg,243.66umol,1 eq) and borane; 2-methylpyridine (52.12 mg,487.32umol,2 eq). After the addition, the reaction solution was stirred at 20 ℃ for 12h. LCMS showed starting material was consumed and the desired MS was found. The reaction mixture was diluted with water (15 mL) and extracted with dichloromethane (3×15 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%;35 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ (2R) -2- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] morpholin-4-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (112.9 mg,139.18umol,57.12% yield, 98.98% purity) as a yellow solid.
Exemplary Synthesis of Compound 157
Compound 157 was prepared in a similar manner to compound 156 using tert-butyl (2R) -2- (p-tolylsulfonyloxymethyl) morpholine-4-carboxylate.
Exemplary Synthesis of Compound 158
Step 1
To a solution of (2S) -2- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] morpholine (140.00 mg,311.42umol,1.71 eq.) in MeOH (3 mL) and HOAc (0.3 mL) was added 2- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] acetaldehyde (70 mg,182.58umol,1 eq.) and borane; 2-methylpyridine (39.06 mg,365.16umol,2 eq). After the addition, the reaction solution was stirred at 20 ℃ for 12h. LCMS showed the desired MS. The reaction mixture was diluted with water (15 mL) and extracted with dichloromethane (3×15 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%;35 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [2- [ (2R) -2- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] morpholin-4-yl ] ethyl ] -1-piperidinyl ] isoindoline-1, 3-dione (94.3 mg,114.86umol,62.91% yield, 99.51% purity) as a yellow solid.
Exemplary Synthesis of Compound 159
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (450 mg,1.63mmol,9.41e-1 eq.) and 4- (2, 2-dimethoxyethyl) piperidine (300 mg,1.73mmol,1 eq.) in DMSO (5 mL) was added DIEA (2.24 g,17.32mmol,3.02mL,10 eq.) at 20deg.C in one portion. The mixture was stirred at 100℃for 16h. LCMS showed the reaction was complete. The mixture was cooled to 20 ℃. The residue was poured into HCl (2M) to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (3X 10 mL). The combined organic phases were washed with brine (2×10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% MeOH/DCM) to give 5- [4- (2, 2-dimethoxyethyl) -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (673 mg,1.57mmol,90.50% yield) as a yellow gum.
Step 2
To a mixture of 5- [4- (2, 2-dimethoxyethyl) -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (350 mg,814.97umol,1 eq.) in THF (5 mL) at 25 ℃ was added HCl (2 m,69.94mL,171.63 eq.) in one portion. The mixture was stirred at 25 ℃ for 2 hours to give a yellow solution. LCMS showed the desired MS. The residue was poured into saturated NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give 2- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] acetaldehyde (290 mg,658.06umol,80.75% yield, 87% purity) as a yellow solid.
Step 3
To a mixture of 5- (1-methylcyclopropoxy) -3- [6- [ (3S) -3-methyl-4- (4-piperidinylmethyl) piperazin-1-yl ] pyrimidin-4-yl ] -1H-indazole (60 mg,129.98umol,1 eq.) and 2- [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindol-5-yl ] -4-piperidinyl ] acetaldehyde (59.80 mg,155.98umol,1.2 eq.) in MeOH (5 mL) at 20 ℃ was added HOAc (0.5 mL) and borane in one portion; 2-methylpyridine (27.81 mg, 259.96. Mu. Mol,2 eq.). The mixture was stirred at 20℃for 16h. LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C1875*30mm*3um; conditions: water (0.225% FA) -ACN; beginning B:0; ending B:35; flow rate: 35mL/min; gradient time: 35min;100% B hold time: 1 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [2- [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] ethyl ] -1-piperidinyl ] isoindoline-1, 3-dione (61.9 mg,73.82umol,56.79% yield, 98.86% purity) as a yellow solid.
Exemplary Synthesis of Compound 160
Step 1
To a solution of tert-butyl 3-methyleneazetidine-1-carboxylate (2.5 g,14.77mmol,1 eq.) was added 9-BBN (0.5M, 29.55mL,1 eq.) at 25 ℃. The reaction mixture was stirred under N2 at 80℃for 1h. After cooling, 4-bromopyridine (2.33 g,14.77mmol,1 eq), pd (dppf) Cl2 (648.59 mg,886.41 mol,0.06 eq), K2CO3 (3.06 g,22.16mmol,1.5 eq), DMF (20 mL), and H2O (5 mL) were added to the reaction. The resulting mixture was heated to 60 ℃ for 12h. LCMS showed the desired MS. After cooling, the reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (3×50 mL). The organic layer was washed with brine (2×50 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 40% ethyl acetate/petroleum ether) to give tert-butyl 3- (4-pyridylmethyl) azetidine-1-carboxylate (2.1 g,8.46mmol,57.24% yield) as a yellow oil.
Step 2
To a solution of tert-butyl 3- (4-pyridylmethyl) azetidine-1-carboxylate (2.1 g,8.46mmol,1 eq.) in EtOH (20 mL) and HOAc (507.85 mg,8.46mmol,483.67uL,1 eq.) was added PtO2 (288.05 mg,1.27mmol,0.15 eq.) at 25 ℃. The mixture was then stirred at 70℃under H2 (50 psi) for 16H. TLC (PE: ea=1:1) showed that the starting material was consumed and a new spot formed. After cooling, the reaction was filtered and the filtrate was concentrated under reduced pressure to give tert-butyl 3- (4-piperidinylmethyl) azetidine-1-carboxylate (2.7 g, crude) as a yellow gum.
Step 3
To a mixture of tert-butyl 3- (4-piperidinylmethyl) azetidine-1-carboxylate (1 g,3.93mmol,1 eq.) and benzyl 4-formylpiperidine-1-carboxylate (972.17 mg,3.93mmol,1 eq.) in MeOH (10 mL) at 25 ℃ was added HOAc (236.08 mg,3.93mmol,224.83ul,1 eq.) in one portion; 2-methylpyridine (841.00 mg,7.86mmol,2 eq.) and HOAc (236.08 mg,3.93mmol,224.83uL,1 eq.). The mixture was stirred at 25℃for 1h. TLC (dichloromethane: methanol=10:1, rf=0.21, pma) showed that the reaction was complete. LCMS showed the desired MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10:1, rf= 0.43,0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give benzyl 4- [ [4- [ (1-tert-butoxycarbonyl azetidin-3-yl) methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (1 g,1.89mmol,48.19% yield, 92% purity) as a yellow oil.
Step 4
To a mixture of benzyl 4- [ [4- [ (1-tert-butoxycarbonylazetidin-3-yl) methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (1 g,2.06mmol,1 eq.) in DCM (10 mL) at 25℃was added TFA (7.70 g,67.53mmol,5mL,32.80 eq.) in one portion. The mixture was stirred at 25℃for 30min. TLC showed the reaction was complete. The residue was poured into aqueous NaHCO3 to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 3), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to give benzyl 4- [ [4- (azetidin-3-ylmethyl) -1-piperidinyl ] methyl ] piperidine-1-carboxylate (800 mg,1.39mmol,67.52% yield, 67% purity) as a yellow oil.
Step 5
To a mixture of 4-bromo-2-fluoro-pyridine (365.17 mg,2.08mmol,1 eq) and 4- [ [4- (azetidin-3-ylmethyl) -1-piperidinyl ] methyl ] piperidine-1-carboxylic acid benzyl ester (800 mg,2.08mmol,1 eq) in DMSO (10 mL) at 100 ℃ was added K2CO3 (1.43 g,10.38mmol,70.14mL,5 eq) in one portion. The mixture was stirred at 100℃for 2h to give a yellow solution. LCMS showed the reaction was complete. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (50 mL). The aqueous phase was extracted with ethyl acetate (50 ml x 4). The combined organic phases were washed with brine (50 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=1:1, rf=0.56, 20g,0% -50% (10 min) ethyl acetate/petroleum ether, 50% (10 min) ethyl acetate/petroleum ether) to give benzyl 4- [ [4- [ [1- (4-bromo-2-pyridinyl) azetidin-3-yl ] methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (830 mg,1.53mmol,73.87% yield) as a yellow oil.
Step 6
To a solution of benzyl 4- [ [4- [ [1- (4-bromo-2-pyridinyl) azetidin-3-yl ] methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (423 mg,781.13umol,1 eq.) and 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl) -1,3, 2-dioxapentaborane (396.72 mg,1.56mmol,2 eq.) in dioxane (10 mL) was added Pd (dppf) Cl2 (57.16 mg,78.11umol,0.1 eq.) and KOAc (229.98 mg,2.34mmol,3 eq.). The mixture was then stirred at 100 ℃ under N2 for 16 hours. TLC (dichloromethane: methanol=10:1, rf=0.5) showed a new spot for the reaction. The reaction mixture was filtered and concentrated under reduced pressure to give [2- [3- [ [1- [ (1-benzyloxycarbonyl-4-piperidinyl) methyl ] -4-piperidinyl ] methyl ] azetidin-1-yl ] -4-pyridinyl ] boronic acid (395 mg, crude) as a brown solid.
Step 7
To a solution of [2- [3- [ [1- [ (1-benzyloxycarbonyl-4-piperidinyl) methyl ] -4-piperidinyl ] methyl ] azetidin-1-yl ] -4-pyridinyl ] boronic acid (399mg, 779.95umol,1 eq.) and 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (250 mg,562.34umol,7.21e-1 eq.) in dioxane (10 mL) and H2O (2 mL) was added Pd (dppf) Cl2 (85.60 mg,116.99umol,0.15 eq.), na2CO3 (248.00 mg,2.34mmol,3 eq.). The mixture was then stirred at 100℃under N2 for 3 hours. TLC (petroleum ether: ethyl acetate=0:1, rf=0.02) showed no starting material and a major new spot of greater polarity was detected. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give benzyl 4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] azetidin-3-yl ] methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylate (175 mg,141.51umol,18.14% yield, 63% purity) as a yellow solid.
Step 8
To a solution of 4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] azetidin-3-yl ] methyl ] -1-piperidinyl ] methyl ] piperidine-1-carboxylic acid benzyl ester (175 mg,224.62umol,1 eq) in MeOH (4 mL) was added HCl/dioxane (4 m,4mL,71.23 eq) and the mixture stirred at 65 ℃ for 1h. The reaction mixture was filtered and concentrated under reduced pressure. The crude product was taken up in TFA (6.16 g,54.02mmol,4.00mL,240.51 eq.) and stirred at 90℃under N2 for 1h. The reaction mixture was filtered and concentrated under reduced pressure to give 5- (1-methylcyclopropoxy) -3- [2- [3- [ [1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] azetidin-1-yl ] -4-pyridinyl ] -1H-indazole (115 mg, crude) as a yellow gum.
Step 9
To a solution of 5- (1-methylcyclopropoxy) -3- [2- [3- [ [1- (4-piperidinylmethyl) -4-piperidinyl ] methyl ] azetidin-1-yl ] -4-pyridinyl ] -1H-indazole (115 mg,223.43 mol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (92.57 mg,335.14 mol,1.5 eq.) in DMSO (5 mL) and DIEA (288.76 mg,2.23mmol,389.17ul,10 eq.). The mixture was stirred at 80℃for 16h. LCMS showed the desired product MS. The resulting product was poured into H2O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na2SO4 and concentrated in vacuo to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN; B%:0% -40%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] azetidin-3-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (8.8 mg,11.30umol,5.06% yield, 99% purity) as a yellow solid.
Exemplary Synthesis of Compound 161
Compound 161 was prepared in an analogous manner to compound 99 starting from tert-butyl 2, 6-diazaspiro [3.3] heptane-2-carboxylate.
Step 1
To a solution of 5- (1-methylcyclopropoxy) -3- [2- [6- (4-piperidinylmethyl) -2, 6-diazaspiro [3.3] hept-2-yl ] -4-pyridinyl ] -1H-indazole (82 mg,178.81 mol,1 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (100 mg,270.73 mol,1.51 eq.) in MeOH (3 mL) and HOAc (0.3 mL) was added borane; 2-methylpyridine (57.38 mg,536.42umol,3 eq). After the addition, the reaction solution was stirred at 25 ℃ for 2h. LCMS showed the starting material was consumed and the desired MS was detected. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%;30 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [2- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -2, 6-diazaspiro [3.3] hept-6-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (45.8 mg,55.53umol,31.06% yield, 98.45% purity) as a yellow solid.
Exemplary Synthesis of Compound 162
Compound 162 was prepared in a similar manner to compound 99 starting from tert-butyl 1, 6-diazaspiro [3.3] heptane-6-carboxylate.
Step 1
To a mixture of 5- (1-methylcyclopropoxy) -3- [2- [6- (4-piperidinylmethyl) -1, 6-diazaspiro [3.3] hept-1-yl ] -4-pyridinyl ] -1H-indazole (80 mg,174.45umol,1 eq.) in MeOH (2 mL) and HOAc (0.2 mL) at 25 ℃ was added borane in one portion; 2-methylpyridine (37.32 mg,348.89umol,2 eq) 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (96.65 mg,261.67umol,1.5 eq) followed by borane addition; 2-methylpyridine (37.32 mg, 348.89. Mu. Mol,2 eq.). The mixture was stirred at 25℃for 12 hours. LCMS showed the reaction was complete. The mixture was poured into H2O (0.5 mL) and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:10% -40%,30 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -1, 6-diazaspiro [3.3] hept-6-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (124.4 mg,151.68umol,86.95% yield, 99% purity) as a pale yellow solid.
Exemplary Synthesis of Compound 163
Compound 163 was prepared in an analogous manner to compound 99 starting from tert-butyl 1, 6-diazaspiro [3.3] heptane-6-carboxylate.
Step 1
To a solution of 5- (1-methylcyclopropoxy) -3- [2- [1- (4-piperidinylmethyl) -1, 6-diazaspiro [3.3] hept-6-yl ] -4-pyridinyl ] -1H-indazole (83 mg,144.95umol,1 eq, TFA) in DCM (5 mL) was added DIEA (93.67 mg,724.74umol,126.23ul,5 eq) and stirred at 20 ℃ for 10min. The mixture was then concentrated. The residue was stirred with a solution of 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (58.89 mg,159.44umol,1.1 eq.) in HOAC (1 mL) and MeOH (10 mL) at 20 ℃ for 20min, followed by the addition of borane; 2-methylpyridine (31.01 mg,289.90umol,2 eq). The mixture was then stirred at 25 ℃ under N2 for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,35 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [6- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -1, 6-diazaspiro [3.3] hept-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (22.2 mg,26.99umol,18.62% yield, 98.73% purity) as a yellow solid.
Exemplary Synthesis of Compound 164
Step 1
To a mixture of tert-butyl 3- (4-piperidinylmethyl) azetidine-1-carboxylate (68.86 mg,270.73umol,1 eq), 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (100 mg,270.73umol,1 eq) in MeOH (10 mL) was added AcOH (1 mL), borane; 2-methylpyridine (57.92 mg,541.46 mol,2 eq.) then the mixture was stirred under N2 at 25℃for 2 hours. LCMS showed complete consumption of starting material and found the desired MS. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% to 30% dichloromethane/methanol) to give tert-butyl 3- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] azetidine-1-carboxylate (133 mg,94.10umol,34.76% yield, 43% purity) as a yellow solid.
Step 2
To a mixture of tert-butyl 3- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] azetidine-1-carboxylate (133 mg,218.84umol,1 eq) in DCM (2 mL) was added TFA (3.08 g,27.01mmol,2mL,123.43 eq), followed by stirring the mixture under N2 at 25 ℃ for 2 hours. TLC (petroleum ether: ethyl acetate=3:1) found a new spot. The reaction mixture was concentrated under reduced pressure to give 5- [4- [ [4- (azetidin-3-ylmethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (100 mg, crude TFA) as a brown oil.
Step 3
To a solution of 5- [4- [ [4- (azetidin-3-ylmethyl) -1-piperidinyl ] methyl ] -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (100 mg,197.00umol,1 eq.) in DMSO (4 mL) was added 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (59.25 mg,197.00umol,1 eq.) DIEA (25.46 mg,197.00umol,34.31uL,1 eq.). The mixture was then stirred at 70 ℃ under N2 for 16 hours. LCMS showed the desired MS. The residue was concentrated under reduced pressure to give a residue. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] azetidin-3-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (8 mg,10.23umol,5.19% yield, 98.7% purity) as a yellow solid.
Exemplary Synthesis of Compound 165
Compound 165 was prepared in a similar manner to compound 99 using tert-butyl 4- [2- (azetidin-3-yl) ethyl ] piperidine-1-carboxylate.
Step 1
To a mixture of tert-butyl 4- (2-bromoethyl) piperidine-1-carboxylate (900 mg,3.08mmol,1 eq.) and dimethyl malonate (488.29 mg,3.70mmol,424.60ul,1.2 eq.) in MeCN (10 mL) was added K2CO3 (1.28 g,9.24mmol,3 eq.) at 25 ℃ in one portion. The mixture was stirred at 90℃for 12 hours. TLC showed the reaction was complete. The mixture was filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 10/1) to give dimethyl 2- [2- (1-tert-butoxycarbonyl-4-piperidinyl) ethyl ] malonate (920 mg,2.68mmol,86.98% yield) as a yellow solid.
Step 2
LAH (305.04 mg,8.04mmol,3 eq.) was added in one portion to a mixture of dimethyl 2- [2- (1-tert-butoxycarbonyl-4-piperidinyl) ethyl ] malonate (920 mg,2.68mmol,1 eq.) in THF (10 mL) at-40 ℃ in N2. The mixture was stirred at-40℃for 4 hours. TLC showed the reaction was complete. Water (0.5 mL) and 10% aqueous NaOH (0.5 mL) were added to the mixture. The aqueous phase was filtered over anhydrous Na2SO4 and concentrated in vacuo to give 4- [ 4-hydroxy-3- (hydroxymethyl) butyl ] piperidine-1-carboxylic acid tert-butyl ester (750 mg,2.61mmol,97.41% yield) as a yellow solid.
Step 3
To a mixture of tert-butyl 4- [ 4-hydroxy-3- (hydroxymethyl) butyl ] piperidine-1-carboxylate (750.00 mg,2.61mmol,1 eq.) and TEA (2.64 g,26.10mmol,3.63mL,10 eq.) in DCM (5 mL) was added at 0deg.C TosCl (2.49 g,13.05mmol,5 eq.) and DMAP (31.88 mg,260.97umol,0.1 eq.) in one portion. The mixture was stirred at 25℃for 12 hours. TLC showed the reaction was complete. The mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 5/1) to give 4- [4- (p-toluenesulfonyloxy) -3- (p-toluenesulfonyloxymethyl) butyl ] piperidine-1-carboxylic acid tert-butyl ester (560 mg,939.97umol,36.02% yield) as a yellow solid.
Step 4
To a mixture of tert-butyl 4- [4- (p-toluenesulfonyloxy) -3- (p-toluenesulfonyloxymethyl) butyl ] piperidine-1-carboxylate (500 mg,839.26umol,1 eq.) and PMBNH2 (172.69 mg,1.26mmol,162.92uL,1.5 eq.) in MeCN (15 mL) was added DIEA (325.40 mg,2.52mmol,438.55uL,3 eq.) at 25℃in one portion. The mixture was stirred at 90℃for 12 hours. LCMS showed the desired MS. The mixture was concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:10% -50%,40 min) to give tert-butyl 4- [2- [1- [ (4-methoxyphenyl) methyl ] azetidin-3-yl ] ethyl ] piperidine-1-carboxylate (150 mg,150.56umol,17.94% yield, 39% purity) as a yellow solid.
Step 5
To a mixture of tert-butyl 4- [2- [1- [ (4-methoxyphenyl) methyl ] azetidin-3-yl ] ethyl ] piperidine-1-carboxylate (20 mg,51.47 mol,1 eq.) in t-BuOH (2 mL) and HOAc (0.1 mL) was added Pd (OH) 2 (10 mg,71.21 mol,1.38 eq.) at 25℃in one portion. The mixture was stirred at 45℃for 12 hours. The mixture was filtered and concentrated in vacuo to give tert-butyl 4- [2- (azetidin-3-yl) ethyl ] piperidine-1-carboxylate (15 mg,45.67umol,88.72% yield, HOAC) as a yellow solid.
Step 6
To a mixture of 5- (1-methylcyclopropoxy) -3- [2- [3- [2- (4-piperidinyl) ethyl ] azetidin-1-yl ] -4-pyridinyl ] -1H-indazole (120 mg,219.94umol,1 eq., TFA) and DIEA (28.43 mg,219.94umol,38.31ul,1 eq.) in MeOH (10 mL) at 25 ℃ was added 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (81.24 mg,219.94umol,1 eq.) and HOAc (1 mL) at once. The mixture was stirred at 25 ℃ for 30min, followed by the addition of (2-methylpyridin-1-ium-1-yl) borohydride (70.58 mg,659.83umol,3 eq.) and the mixture stirred at 25 ℃ for 11.5 hours. The mixture was concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 10% -50%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [2- [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] azetidin-3-yl ] ethyl ] -1-piperidinyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (57.2 mg,70.98umol,32.27% yield, 97.40% purity) as a yellow solid.
Exemplary Synthesis of Compound 166
Step 1
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindoline-1, 3-dione (300 mg, 541.98. Mu.l, 1.28 eq., TFA) and tert-butyl (3S) -3- (p-toluenesulfonyloxymethyl) pyrrolidine-1-carboxylate (150 mg, 422.00. Mu.mol, 1 eq.) in CH3CN (5 mL) was added DIEA (272.70 mg,2.11mmol, 367.52. Mu.L, 5 eq.) and KI (140.10 mg, 844.01. Mu.mol, 2 eq.). After the addition, the reaction solution mixture was stirred at 80 ℃ for 12h. LCMS showed the desired MS. After cooling, the reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (3×10 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% methanol in dichloromethane) to give (3R) -3- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] pyrrolidine-1-carboxylic acid tert-butyl ester (320 mg, crude) as a yellow solid.
Step 2
To a solution of tert-butyl (3R) -3- [ [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] pyrrolidine-1-carboxylate (160 mg,256.92umol,1 eq.) in DCM (5 mL) was added TFA (29.29 mg,256.92umol,19.02ul,1 eq.). After the addition, the reaction solution mixture was stirred at 25 ℃ for 1h. LCMS showed starting material was consumed and the desired MS was found. The reaction mixture was concentrated under reduced pressure to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (3S) -pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (165 mg, crude TFA) as a yellow gum. The crude product was used directly in the next step.
Step 3
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (3S) -pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (135 mg,212.04umol,1.28 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (50 mg,166.26umol,1 eq) in DMSO (1.5 mL) was added DIEA (107.44 mg,831.28umol,144.79ul,5 eq). After the addition, the reaction solution was stirred at 90 ℃ for 16h. LCMS showed the desired MS. After cooling, the reaction mixture was diluted with ethyl acetate (10 mL) and washed with brine (2×5 mL). The organic layer was concentrated under reduced pressure. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%;40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (3R) -1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (18.5 mg,23.21umol,13.96% yield, 98.71% purity) as a yellow solid.
Exemplary Synthesis of Compound 167
Compound 167 was prepared in a similar manner to compound 166 starting from tert-butyl (3R) -3- (p-tolylsulfonyloxymethyl) pyrrolidine-1-carboxylate.
Step 1
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (3R) -pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (400 mg,462.58 mol,2.32 eq, 3 TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (60 mg,199.51 mol,1 eq) in DMSO (5 mL) was added DIPEA (206.28 mg,1.60mmol,278.00ul,8 eq) at 20 ℃ in one portion. The mixture was stirred at 80℃for 16h. LCMS showed the desired MS. The mixture was cooled to 20 ℃ and concentrated under reduced pressure at 20 ℃. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; conditions: water (0.225% FA) -ACN; start B:0 end B:30; flow rate: 25mL/min; gradient time: 40min;100% B hold time: 3 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (3S) -1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] pyrrolidin-3-yl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (15.7 mg,19.28umol,9.66% yield, 96.63% purity) as a yellow solid.
Exemplary Synthesis of Compound 168
Step 1
To a mixture of tert-butyl (3S) -3-methylpiperazine-1-carboxylate (1 g,4.99mmol,1 eq.) and 3-bromoprop-1-yne (742.47 mg,4.99mmol,538.02ul,80% purity, 1 eq.) in MeCN (10 mL) was added DIEA (1.29 g,9.99mmol,1.74mL,2 eq.) at 25℃in one portion. The mixture was stirred at 25℃for 120min. TLC (petroleum ether: ethyl acetate=1/1) showed the reaction was complete. The mixture was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (15 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=3/1) to give (3S) -3-methyl-4-prop-2-ynyl-piperazine-1-carboxylic acid tert-butyl ester (1.0 g,4.20mmol,84.04% yield) as a pale yellow liquid.
Step 2
To a mixture of benzyl 4- (hydroxymethyl) piperidine-1-carboxylate (4 g,16.04mmol,1 eq), DMAP (392.03 mg,3.21mmol,0.2 eq) and TEA (4.87 g,48.13mmol,6.70mL,3 eq) in DCM (50 mL) was added 4-methylbenzenesulfonyl chloride (6.12 g,32.09mmol,2 eq) at 0deg.C. The mixture was stirred at 25℃for 2 hours. TLC (petroleum ether: ethyl acetate=3/1) showed that a new spot formed and the desired compound was detected by LCMS. The mixture was poured into water (50 mL). The aqueous phase was extracted with DCM (30 ml x 2). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=3/1) to give benzyl 4- (p-toluenesulfonyloxymethyl) piperidine-1-carboxylate (5.7 g,14.13mmol,88.05% yield) as a grey solid.
Step 3
To a mixture of benzyl 4- (p-toluenesulfonyloxymethyl) piperidine-1-carboxylate (2 g,4.96mmol,1 eq.) in DMF (20 mL) and H2O (2 mL) at 25℃was added NaN3 (500 mg,7.69mmol,1.55 eq.) in one portion. The mixture was stirred at 80℃for 8 hours. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃ and poured into water (50 mL). The aqueous phase was extracted with DCM (20 ml x 3). The combined organic phases were washed with brine (20 ml x 3), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was used in the next step without further purification. The product 4- (azidomethyl) piperidine-1-carboxylic acid benzyl ester (1.1 g,4.01mmol,80.90% yield) was a pale yellow liquid.
Step 4
A mixture of benzyl 4- (azidomethyl) piperidine-1-carboxylate (900 mg,3.28mmol,1 eq), (3S) -3-methyl-4-prop-2-ynyl-piperazine-1-carboxylate (781.91 mg,3.28mmol,1 eq), cuSO4.5H2O (409.60 mg,1.64mmol,0.5 eq) and sodium ascorbate (454.98 mg,2.30mmol,0.7 eq) in t-BuOH (5 mL) and H2O (5 mL) was stirred at 25℃for 12H. LCMS showed the reaction was complete. The reaction mixture was poured into H2O (10 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (20 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. The residue was purified by silica gel chromatography (dichloromethane/methanol=10/1) to give (3S) -4- [ [1- [ (1-benzyloxycarbonyl-4-piperidinyl) methyl ] triazol-4-yl ] methyl ] -3-methyl-piperazine-1-carboxylic acid tert-butyl ester (1.5 g,2.93mmol,89.18% yield) as a brown solid.
Step 5
To a solution of tert-butyl (3S) -4- [ [1- [ (1-benzyloxycarbonyl-4-piperidinyl) methyl ] triazol-4-yl ] methyl ] -3-methyl-piperazine-1-carboxylate (240 mg,468.16umol,1 eq.) in MeOH (10 mL) was added nh3.h2o (117.19 mg,936.32umol,128.78ul,28% purity, 2 eq.) and Pd/C (468.16 umol,10% purity, 1 eq.) followed by stirring the mixture at 25 ℃ under H2 (15 psi) for 12 hours. LCMS showed the reaction was complete. The mixture was filtered and concentrated in vacuo. The crude product was used in the next step without further purification. The product (3S) -3-methyl-4- [ [1- (4-piperidinylmethyl) triazol-4-yl ] methyl ] piperazine-1-carboxylic acid tert-butyl ester (170 mg,449.13umol,95.93% yield) was a grey solid.
Step 6
To a mixture of tert-butyl (3S) -3-methyl-4- [ [1- (4-piperidinylmethyl) triazol-4-yl ] methyl ] piperazine-1-carboxylate (170 mg,449.13umol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (124.06 mg,449.13umol,1 eq.) in DMSO (3 mL) was added DIEA (290.23 mg,2.25mmol,391.15uL,5 eq.) in one portion. The mixture was stirred at 100℃for 2 hours. TLC (dichloromethane: methanol=10/1) showed the reaction was complete. The mixture was cooled to 25 ℃ and poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (15 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane: methanol=10/1) to give (3S) -4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] triazol-4-yl ] methyl ] -3-methyl-piperazine-1-carboxylic acid tert-butyl ester (100 mg,152.82umol,34.03% yield, 97% purity) as a yellow solid.
Step 7
To a mixture of (3S) -4- [ [1- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] triazol-4-yl ] methyl ] -3-methyl-piperazine-1-carboxylic acid tert-butyl ester (100 mg,157.55umol,1 eq.) in DCM (1 mL) was added TFA (1.54 g,13.51mmol,1mL,85.73 eq.) in one portion. The mixture was stirred at 25℃for 1 hour. TLC (methanol: dichloromethane=1/10) showed the reaction was complete. The mixture was concentrated under reduced pressure at 35 ℃. The crude product was used in the next step without further purification. The product 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] triazol-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (75 mg,138.89umol,88.15% yield, 99% purity) was a pale yellow oil.
Step 8
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] triazol-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (75 mg,140.29umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (42.19 mg,140.29umol,1 eq.) in DMSO (3 mL) was added DIEA (181.31 mg,1.40mmol,244.36ul,10 eq.) in one portion. The mixture was stirred at 84℃for 3 hours. LCMS showed 8% residue of reactant EB153-506-P1 and produced 44% of the desired product. The mixture was cooled to 25 ℃ and poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (15 ml x 2). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex Luna C18 75X 30mm X3 um; mobile phase: [ water (10 mM NH4HCO 3) -ACN ]; B%:0% -70%,35 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] triazol-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (30 mg,37.10umol,26.45% yield, 98.8% purity) as a yellow solid.
Exemplary Synthesis of Compound 169
Step 1
To 4- (bromomethyl) benzaldehyde (2 g,10.05mmol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester; to a mixture of hydrochloride (2.24 g,10.05mmol,1 eq.) in MeCN (15 mL) was added K2CO3 (4.17 g,30.15mmol,3 eq.) in one portion. The mixture was stirred at 25℃for 4 hours. LCMS showed the reaction was complete. The mixture was filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=4/1) to give tert-butyl 4- [ (4-formylphenyl) methyl ] piperazine-1-carboxylate (2.7 g,8.87mmol,88.26% yield) as a pale yellow oil.
Step 2
To a mixture of tert-butyl 4- [ (4-formylphenyl) methyl ] piperazine-1-carboxylate (2.5 g,8.21mmol,1 eq.) in HOAc (1.5 mL) and MeOH (15 mL) was added benzyl piperazine-1-carboxylate (2.17 g,9.86mmol,1.90mL,1.2 eq.) at 25 ℃ in one portion followed by borane; 2-methylpyridine (1.76 g,16.43mmol,2 eq.). The mixture was stirred at 25℃for 12 hours. LCMS showed the reaction was complete. The mixture was concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1/1) to give benzyl 4- [ [4- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] phenyl ] methyl ] piperazine-1-carboxylate (3.5 g,6.88mmol,83.78% yield) as a grey solid.
Step 3
To a solution of benzyl 4- [ [4- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] phenyl ] methyl ] piperazine-1-carboxylate (300 mg,589.80 mol,1 eq) in DCM (2 mL) was added TFA (1.54 g,13.51mmol,1mL,22.90 eq), and the mixture was stirred at 25℃for 1 hour. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃ and concentrated under reduced pressure at 40 ℃. The crude product was used in the next step without further purification. The product 4- [ [4- (piperazin-1-ylmethyl) phenyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (220 mg,527.74umol,89.48% yield, 98% purity) was as a pale yellow liquid.
Step 4
To a mixture of benzyl 4- [ [4- (piperazin-1-ylmethyl) phenyl ] methyl ] piperazine-1-carboxylate (110 mg,269.25umol,1 eq.) and 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (74.37 mg,269.25umol,1 eq.) in DMSO (2 mL) was added DIEA (174.00 mg,1.35mmol,234.50uL,5 eq.) in one portion. The mixture was stirred at 100℃for 12 hours. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃ and poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (15 ml x 2). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (dichloromethane/methanol=10/1) to give benzyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] phenyl ] methyl ] piperazine-1-carboxylate (100 mg,150.43umol,55.87% yield) as a pale yellow solid.
Step 5
To a mixture of benzyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] phenyl ] methyl ] piperazine-1-carboxylate (100 mg,150.43umol,1 eq.) in TFA (2 mL), the mixture was stirred at 80 ℃ for 1 hour. TLC (dichloromethane: methanol=10/1) showed the reaction was complete. The mixture was cooled to 25 ℃ and concentrated under reduced pressure at 60 ℃. The crude product was used in the next step without further purification. The product 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (piperazin-1-ylmethyl) phenyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (75 mg,126.79umol,84.28% yield, 89.7% purity) was a pale yellow oil.
Step 6
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (piperazin-1-ylmethyl) phenyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (37 mg,69.73umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (20.97 mg,69.73umol,1 eq.) in DMSO (2 mL) was added DIEA (90.12 mg,697.30umol,121.46ul,10 eq.) in one portion. The mixture was stirred at 83℃for 3 hours. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃ and poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 2). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -30%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] phenyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (9.5 mg,10.98umol,15.75% yield, 97.19% purity, FA) as a pale yellow solid.
Exemplary Synthesis of Compound 170
Compound 170 was prepared in a similar manner to compound 169 starting from 3- (bromomethyl) benzaldehyde.
Step 1
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- (piperazin-1-ylmethyl) phenyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (100 mg,188.46 mol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (56.68 mg,188.46 mol,1 eq.) in DMSO (3.5 mL) was added DIEA (371.00 mg,2.87mmol,500.00ul,15.23 eq.). After the addition, the mixture was stirred at 80 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (30 mL). The aqueous phase was extracted with ethyl acetate (30 ml x 3). The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -30%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] phenyl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (19.2 mg,24.03umol,12.75% yield, 99.50% purity) as a yellow solid.
Exemplary Synthesis of Compound 171
Compound 171 was prepared in an analogous manner to compound 169 using benzyl 4- [ [4- (4-piperidinylmethyl) phenyl ] methyl ] piperidine-1-carboxylate.
Step 1
To a solution of tert-butyl 4-methylenepiperidine-1-carboxylate (3.84 g,19.44mmol,1.1 eq.) 9-BBN (0.5M, 42.42mL,1.2 eq.) was added and the mixture stirred under N2 at 70℃for 1h. The solution was then cooled to 30 ℃ and 1-bromo-4-iodo-benzene (5 g,17.67mmol,1 eq.) was added followed by K2CO3 (7.33 g,53.02mmol,3 eq.), DMF (40 mL), water (4 mL) and Pd (dppf) Cl2 (1.94 g,2.65mmol,0.15 eq.). The solution was heated to 70 ℃ under N2 and held for 3 hours. LCMS showed the desired MS values and TLC (petroleum ether: ethyl acetate=7:1, rf=0.66, uv=254 nm) showed the formation of new spots. Water (50 mL) was added to the reaction mixture and extracted with EA (80 mL. Times.3). The combined organic layers were washed with brine (80 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel (0% -7% ethyl acetate/petroleum ether) to give tert-butyl 4- [ (4-bromophenyl) methyl ] piperidine-1-carboxylate (2 g,4.65mmol,26.29% yield, 82.3% purity) as a pale yellow oil.
Step 2
To a solution of benzyl 4-methylenepiperidine-1-carboxylate (783.41 mg,3.39mmol,1.2 eq.) was added 9-borobicyclo [3.3.1] nonane (0.5 m,6.77ml,1.2 eq.) and the mixture stirred under N2 at 80 ℃ for 1h. The solution was then cooled to 30℃and tert-butyl 4- [ (4-bromophenyl) methyl ] piperidine-1-carboxylate (1 g,2.82mmol,1 eq.) was added followed by K2CO3 (1.17 g,8.47mmol,3 eq.), DMF (20 mL), water (5 mL) and Pd (dppf) Cl2 (165.23 mg,225.81umol,0.08 eq.). The solution was heated to 80 ℃ under N2 and held for 15 hours. LCMS showed about 38% of the desired compound and TLC (petroleum ether: ethyl acetate=10:1, I2, rf=0.28) showed that a new spot formed and the starting material was completely consumed. To the reaction mixture was added 50mL of water and extracted with EA (80 mL x 3). The combined organic layers were washed with 80mL of brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel (0% -7% ethyl acetate/petroleum ether) to give benzyl 4- [ [4- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] phenyl ] methyl ] piperidine-1-carboxylate (650 mg,1.18mmol,41.95% yield, 91.6% purity) as a yellow solid.
Step 3
To a solution of benzyl 4- [ [4- [ (1-tert-butoxycarbonyl-4-piperidinyl) methyl ] phenyl ] methyl ] piperidine-1-carboxylate (650 mg,1.29mmol,1 eq.) in DCM (10 mL) was added HCl/EtOAc (4M, 5mL,15.47 eq.) and the mixture was then stirred at 25℃for 1 hour. LCMS showed the desired MS value and the starting material was completely consumed. The reaction mixture was concentrated under reduced pressure to give benzyl 4- [ [4- (4-piperidinylmethyl) phenyl ] methyl ] piperidine-1-carboxylate (550 mg,1.24mmol,96.03% yield, HCl) as a yellow solid.
Step 4
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- (4-piperidinylmethyl) phenyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (260.57 mg,492.90umol,2.47 eq) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (60 mg,199.51umol,1 eq) in DMSO (3 mL) was added DIEA (206.28 mg,1.60mmol,278.00ul,8 eq), and the mixture was stirred at 90 ℃ for 4 hours. LCMS showed the desired MS value and the starting material was completely consumed. No post-treatment is required. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 20% -70%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] phenyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (10.5 mg,13.19umol,6.61% yield, 99.57% purity) as a yellow solid.
Exemplary Synthesis of Compound 172
Compound 172 was prepared in an analogous manner to compound 169 using benzyl 4- [ [6- (piperazin-1-ylmethyl) pyridazin-3-yl ] methyl ] piperazine-1-carboxylate.
Step 1
To a solution of methyl 6-methylpyridazine-3-carboxylate (5.5 g,36.15mmol,1 eq.) in DMF (200 mL) was added NBS (7.72 g,43.38mmol,1.2 eq.) and AIBN (629.21 mg,3.83mmol,0.106 eq.). The mixture was stirred at 80℃for 2 hours. LC-MS (EB 134-925-P1C) showed about 27% reactant 1 residual. Several new peaks were shown on LC-MS and about 22% of the desired compound was detected. The reaction mixture was diluted with water (200 mL x 5) and extracted with EA (300 mL). The combined organic layers were washed with brine (200 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by column chromatography on silica gel (column height: 80g,100-200 mesh silica gel, 0% -20% (25 min) ethyl acetate/petroleum ether, 20% (10 min) ethyl acetate/petroleum ether) to give methyl 6- (bromomethyl) pyridazine-3-carboxylate (740 mg,3.20mmol,8.86% yield) as a yellow solid.
Step 2
To a solution of methyl 6- (bromomethyl) pyridazine-3-carboxylate (300 mg,1.30mmol,1 eq.) in THF (3 mL) was added DIEA (503.44 mg,3.90mmol,678.49uL,3 eq.) and tert-butyl piperazine-1-carboxylate (483.67 mg,2.60mmol,2 eq.). The mixture was stirred at 60℃for 16 hours. LC-MS (EB 134-926-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 93% of the desired compound was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent. The crude product was purified by column chromatography on silica gel (column height: 12g,100-200 mesh silica gel, 0% -40% (15 min) ethyl acetate/petroleum ether, 40% (10 min) ethyl acetate/petroleum ether) to give methyl 6- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] pyridazine-3-carboxylate (380 mg,1.13mmol,87.00% yield) as a white solid.
Step 3
A solution of methyl 6- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] pyridazine-3-carboxylate (380 mg,1.13mmol,1 eq.) in THF (20 mL) was cooled to 0deg.C. Diisobutylaluminum hydride (1M, 2.26mL,2 eq.) was slowly added over 30 minutes. The mixture was stirred at 25℃for 4h. LC-MS (EB 134-928-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 23% of the desired compound was detected. The reaction mixture was diluted with NH4Cl (30 mL). The reaction solution was filtered to remove insoluble material and extracted with EA (30 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give tert-butyl 4- [ (6-formylpyridazin-3-yl) methyl ] piperazine-1-carboxylate (260 mg,848.68umol,75.13% yield) as a yellow solid.
Step 4
To a solution of tert-butyl 4- [ (6-formylpyridazin-3-yl) methyl ] piperazine-1-carboxylate (260 mg,848.68umol,1 eq.) in MeOH (6 mL) and AcOH (0.6 mL) was added borane; 2-methylpyridine (453.88 mg,4.24mmol,5 eq.) and benzyl piperazine-1-carboxylate (280.40 mg,1.27mmol,245.97ul,1.5 eq.). The mixture was stirred at 25℃for 2 hours. LC-MS (EB 134-930-P1A) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 76% of the desired compound was detected. TLC (SiO 2, DCM/meoh=10/1, rf=0.24) indicated that a major new spot of greater polarity was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent. The crude product was purified by column chromatography on silica gel (column height: 12g,100-200 mesh silica gel, 0% -10% (15 min) MeOH/DCM) to give benzyl 4- [ [6- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] pyridazin-3-yl ] methyl ] piperazine-1-carboxylate (330 mg,646.26umol,76.15% yield) as a yellow solid.
Step 5
To a solution of benzyl 4- [ [6- [ (4-tert-butoxycarbonylpiperazin-1-yl) methyl ] pyridazin-3-yl ] methyl ] piperazine-1-carboxylate (150 mg,293.76umol,1 eq.) in DCM (2.5 mL) was added TFA (770.00 mg,6.75mmol,0.5mL,22.99 eq.). The mixture was stirred at 25℃for 0.5 h. TLC (DCM/meoh=10/1, rf=0) indicated no residue of reactant 1 and a major new spot of greater polarity was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent to give benzyl 4- [ [6- (piperazin-1-ylmethyl) pyridazin-3-yl ] methyl ] piperazine-1-carboxylate (150 mg,248.79umol,84.69% yield, 87% purity, TFA) as a yellow gum.
Step 6
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [6- (piperazin-1-ylmethyl) pyridazin-3-yl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (100 mg,154.65umol,1 eq, TFA) in DMSO (3 mL) was added DIEA (199.88 mg,1.55mmol,269.37uL,10 eq) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (46.51 mg,154.65umol,1 eq). The mixture was stirred at 80℃for 2 hours. LC-MS (EB 134-936-P1B) showed no residue of reactant 1. Several new peaks were shown on LC-MS and about 53% of the desired compound was detected. The resulting product was filtered to remove insoluble materials. The impure product was purified by preparative HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN;: 0% -35%,40 min) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [6- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] pyridazin-3-yl ] methyl ] piperazin-1-yl ] isoindoline-1, 3-dione (33.8 mg,42.36umol,27.39% yield, 99.86% purity) as a yellow solid.
Exemplary Synthesis of Compound 173
Step 1
To a solution of 4-bromo-2-fluoropyridine (600 mg,3.41mmol,1 eq.) in DMSO (20 mL) was added tert-butyl 4- [ [4- (4-piperidylmethyl) phenyl ] methyl ] piperidine-1-carboxylate (1.5 g,4.03mmol,1.18 eq.) and K2CO3 (1.41 g,10.23mmol,3 eq.). After the addition, the reaction solution was stirred at 90 ℃ for 12h. TLC (petroleum ether: ethyl acetate=5:1) showed the reaction was complete. After cooling, the reaction mixture was filtered, and the filtrate was diluted with ethyl acetate (50 mL) and washed with brine (3×30 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether) to give tert-butyl 4- [ [4- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methyl ] phenyl ] methyl ] piperidine-1-carboxylate (1.4 g,2.35mmol,68.84% yield, 88.6% purity) as a white solid.
Step 2
To a solution of tert-butyl 4- [ [4- [ [1- (4-bromo-2-pyridinyl) -4-piperidinyl ] methyl ] phenyl ] methyl ] piperidine-1-carboxylate (400 mg,756.83 mmoles, 1 eq.) in dioxane (10 mL) was added Pin2B2 (384.37 mg,1.51mmol,2 eq.), pd (dppf) Cl2 (55.38 mg,75.68 mmoles, 0.1 eq.) and KOAc (222.83 mg,2.27mmol,3 eq.). After the addition, the reaction mixture was stirred at 100 ℃ under N2 for 12h. LCMS showed the starting material was consumed and the desired MS formed. After cooling, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give tert-butyl 4- [ [4- [ [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] -4-piperidinyl ] methyl ] phenyl ] methyl ] piperidine-1-carboxylate (800 mg, crude) as a black oil. The crude product was used directly in the next step.
Step 3
To a solution of tert-butyl 4- [ [4- [ [1- [4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2-pyridinyl ] -4-piperidinyl ] methyl ] phenyl ] methyl ] piperidine-1-carboxylate (800 mg,1.39mmol,4.12 eq.) in dioxane (5 mL) and H2O (1 mL) was added 2- [ [ 3-iodo-5- (1-methylcyclopropoxy) indazol-1-yl ] methoxy ] ethyl-trimethyl-silane (150 mg,337.55umol,1 eq.), na2CO3 (107.33 mg,1.01mmol,3 eq.) and Pd (dppf) Cl2 (17.29 mg,23.63umol,0.07 eq.). After addition, the reaction mixture was stirred at 90 ℃ under N2 for 12h. LCMS showed the reaction was complete. TLC (petroleum ether: ethyl acetate=5:1) showed several new spots. After cooling, the reaction mixture was filtered to remove insoluble materials, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether) to give tert-butyl 4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] phenyl ] methyl ] piperidine-1-carboxylate (140 mg,172.69umol,51.16% yield, 94.5% purity) as a pale yellow gum.
Step 4
To a solution of tert-butyl 4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1- (2-trimethylsilylethoxymethyl) indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] phenyl ] methyl ] piperidine-1-carboxylate (140 mg,182.74umol,1 eq) in MeOH (2 mL) was added HCl/EtOAc (4 m,2mL,43.78 eq). After the addition, the reaction solution was stirred at 65 ℃ for 0.5 hours. LCMS showed the reaction was complete. After cooling, the reaction solution was concentrated under reduced pressure to give 5- (1-methylcyclopropoxy) -3- [2- [4- [ [4- (4-piperidinylmethyl) phenyl ] methyl ] -1-piperidinyl ] -4-pyridinyl ] -1H-indazole (110 mg, crude, HCl) as a pale yellow solid. The crude product was used directly in the next step.
Step 5
To a solution of 5- (1-methylcyclopropoxy) -3- [2- [4- [ [4- (4-piperidinylmethyl) phenyl ] methyl ] -1-piperidinyl ] -4-pyridinyl ] -1H-indazole (110 mg,192.25 mol,1 eq, HCl) in DMSO (3 mL) was added 2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-isoindoline-1, 3-dione (53 mg,191.88umol,1 eq) and DIEA (123.99 mg,959.38umol,167.11ul,5 eq). After the addition, the reaction solution was stirred at 100 ℃ for 12 hours. LCMS showed the reaction was complete. After cooling, the reaction mixture was diluted with ethyl acetate (20 mL) and washed with brine (10 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 72% ethyl acetate/petroleum ether) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] methyl ] phenyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (40.6 mg,49.46umol,25.78% yield, 96.48% purity) as a yellow solid.
Exemplary Synthesis of Compound 174
Compound 174 was prepared in a similar manner to compound 171 starting from 1-bromo-3-iodo-benzene.
Step 1
To a solution of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- (4-piperidinylmethyl) phenyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (90 mg,170.25 mol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (51.20 mg,170.25 mol,1 eq.) in DMSO (3 mL) was added DIEA (371.00 mg,2.87mmol,0.5mL,16.86 eq.). After addition, the mixture was stirred at 90 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex Luna C, 18, 75, 30mm 3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:30% -60%,35 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [3- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] phenyl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (14.7 mg,18.17umol,10.67% yield, 98% purity) as a yellow solid.
Exemplary Synthesis of Compound 175
Step 1
To 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (200 mg,541.46 mol,1 eq.) 2, 6-diazaspiro [3.3] heptane-2-carboxylic acid tert-butyl ester; to a mixture of oxalic acid (158.07 mg,324.88umol,0.6 eq.) in MeOH (10 mL) was added AcOH (1 mL), borane; 2-methylpyridine (57.92 mg,541.46 mol,1 eq.) then the mixture was stirred at 25℃under N2 for 12 hours. LCMS showed complete consumption of starting material and found the desired MS. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (0% to 30% dichloromethane/methanol) to give tert-butyl 6- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -2, 6-diazaspiro [3.3] heptane-2-carboxylate (560 mg, crude) as a yellow gum.
Step 2
To a mixture of tert-butyl 6- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -2, 6-diazaspiro [3.3] heptane-2-carboxylate (300 mg,543.84umol,1 eq) in DCM (2 mL) was added TFA (1.65 g,14.47mmol,1.07mL,26.61 eq), and the mixture was stirred at 25 ℃ under N2 for 2 hours. TLC (dichloromethane: methanol=10:1) showed complete consumption of starting material and found a new main point. The reaction mixture was concentrated under reduced pressure to give 5- [4- (2, 6-diazaspiro [3.3] hept-2-ylmethyl) -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (700 mg, crude, TFA) as a brown gum.
Step 3
To a mixture of 5- [4- (2, 6-diazaspiro [3.3] hept-2-ylmethyl) -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (64.60 mg,143.07umol,1 eq), 1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (54 mg,143.07umol,1 eq) in MeOH (10 mL) was added AcOH (1 mL), borane; 2-methylpyridine (30.61 mg,286.14umol,2 eq.) then the mixture was stirred at 25℃under N2 for 12 hours. LCMS showed complete consumption of starting material and found the desired MS. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [2- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] -2, 6-diazaspiro [3.3] hept-6-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (22.2 mg,27.03umol,18.90% yield, 99% purity) as a yellow solid.
Exemplary Synthesis of Compound 176
Compound 176 was prepared in an analogous manner to compound 175 starting from tert-butyl 1, 6-diazaspiro [3.3] heptane-6-carboxylate.
Step 1
To a mixture of 5- [4- (1, 6-diazaspiro [3.3] hept-1-ylmethyl) -1-piperidinyl ] -2- (2, 6-dioxo-3-piperidinyl) isoindoline-1, 3-dione (77.76 mg,172.21 mol,1 eq), 1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (65 mg,172.21 mol,1 eq) in MeOH (10 mL) was added AcOH (1 mL) and borane; 2-methylpyridine (18.42 mg,172.21umol,1 eq.) the mixture was then stirred at 25℃under an atmosphere of N2 for 16 hours. LCMS showed complete consumption of starting material and found the desired MS. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reverse phase HPLC (column 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [6- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] -1, 6-diazaspiro [3.3] hept-1-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (19.6 mg,23.15umol,13.44% yield, 96% purity) as a yellow solid.
Exemplary Synthesis of Compound 177
Step 1
To tert-butyl 2-formyl-7-azaspiro [3.5] nonane-7-carboxylate (500 mg,1.97mmol,1 eq.) piperazine-1-carboxylate (434.73 mg,1.97mmol,381.34ul,1 eq.) borane; to a mixture of 2-methylpyridine (633.31 mg,5.92mmol,3 eq.) in MeOH (10 mL) was added AcOH (2 mL) borane; 2-methylpyridine (633.31 mg,5.92mmol,3 eq.) then the mixture was stirred at 25℃under an atmosphere of N2 for 16 hours. LCMS showed complete consumption of starting material and found the desired MS. TLC (petroleum ether: ethyl acetate=0:1) showed several new spots. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 80% ethyl acetate/petroleum ether) to give the compound tert-butyl 2- [ (4-benzyloxycarbonyl piperazin-1-yl) methyl ] -7-azaspiro [3.5] nonane-7-carboxylate (933 mg,1.53mmol,77.48% yield, 75% purity) as a colourless gum.
Step 2
To a mixture of tert-butyl 2- [ (4-benzyloxycarbonyl piperazin-1-yl) methyl ] -7-azaspiro [3.5] nonane-7-carboxylate (200 mg,437.06umol,1 eq) in DCM (3 mL) was added TFA (4.62 g,40.52mmol,3mL,92.71 eq), and the mixture was stirred at 25℃under an atmosphere of N2 for 1 hour. TLC (petroleum ether: ethyl acetate=0:1) showed a new spot. The reaction mixture was concentrated under reduced pressure to give benzyl 4- (7-azaspiro [3.5] non-2-ylmethyl) piperazine-1-carboxylate (200 mg, crude, TFA) as a yellow gum.
Step 3
To a mixture of benzyl 4- (7-azaspiro [3.5] non-2-ylmethyl) piperazine-1-carboxylate (156 mg,436.38umol,1 eq), 1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (161.19 mg,436.38umol,1 eq) in MeOH (10 mL) was added AcOH (2 mL), borane; 2-methylpyridine (140.03 mg,1.31mmol,3 eq.) then the mixture was stirred at 25℃under an atmosphere of N2 for 16 hours. LCMS showed complete consumption of starting material and found the desired MS. TLC (dichloromethane: methanol=10:1) showed several new spots. The resulting product was poured into H2O (10 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (50 mL), dried over anhydrous Na2SO4, and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (0% to 30% methanol/dichloromethane) to give benzyl 4- [ [7- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -7-azaspiro [3.5] non-2-yl ] methyl ] piperazine-1-carboxylate (233 mg,219.61umol,50.33% yield, 67% purity) as a yellow gum.
Step 4
A mixture of 4- [ [7- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -1, 3-dioxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -7-azaspiro [3.5] non-2-yl ] methyl ] piperazine-1-carboxylic acid benzyl ester (223 mg,313.70 mol,1 eq), TFA (4.62 g,40.52mmol,3mL,129.16 eq) in TFA (4.62 g,40.52mmol,3mL,129.16 eq) was then stirred at 70℃under an atmosphere of N2 for 1 hour. TLC (dichloromethane: methanol=10:1) showed complete consumption of starting material and a new spot was found. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 30% dichloromethane/methanol) to give 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [ 1-oxo-1- (4-piperidinylmethylimino) -1, 4-thiazinan-4-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (200 mg, crude TFA) as a yellow gum.
Step 5
To a mixture of 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [2- (piperazin-1-ylmethyl) -7-azaspiro [3.5] non-7-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (76.71 mg,133.00umol,1 eq), 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (40 mg,133.00umol,1 eq), DIEA (17.19 mg,133.00umol,23.17ul,1 eq) in DMSO (5 mL) was added DIEA (17.19 mg,133.00umol,23.17ul,1 eq) followed by stirring the mixture at 100 ℃ under an N2 atmosphere for 12 hours. LCMS showed complete consumption of starting material and found the desired MS. The reaction mixture was filtered and concentrated under reduced pressure. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 2- (2, 6-dioxo-3-piperidinyl) -5- [4- [ [2- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -7-azaspiro [3.5] non-7-yl ] methyl ] -1-piperidinyl ] isoindoline-1, 3-dione (40.8 mg,48.03umol,36.11% yield, 99% purity) as a yellow solid.
Exemplary Synthesis of Compound 178
Step 1
At 20 ℃ at N 2 Pd (dba) was added in one portion to a mixture of 2-chloro-4-methyl-5-nitro-pyridine (5 g,28.97mmol,1 eq.) and 1-methylcyclopropanol (2.09 g,28.97mmol,1 eq.) in toluene (10 mL) 2 (333.21 mg,579.48umol,0.02 eq.) Cs 2 CO 3 (11.33 g,34.77mmol,1.2 eq.) and BINAP (1.08 g,1.74mmol,0.06 eq.). The mixture was heated to 110 ℃ and stirred for 3 hours. LCMS showed the desired MS. The mixture was cooled to 20 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether) to give 4-methyl-2- (1-methylcyclopropoxy) -5-nitro-pyridine (5.1 g,24.49mmol,84.54% yield) as a yellow oil.
Step 2
At 20 ℃ at N 2 Pd/C (500 mg,24.49mmol,10% purity, 1 eq.) and ammonium formate (18.54 g,293.93mmol,12 eq.) were added in one portion to a mixture of 4-methyl-2- (1-methylcyclopropoxy) -5-nitro-pyridine (5.1 g,24.49mmol,1 eq.) in EtOH (10 mL). The mixture was stirred at 20℃for 1h to give a black solution. TLC showed the reaction was complete. The mixture was filtered through a pad of silica gel, with EtOAc (3×200 mL), and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% ethyl acetate/petroleum ether) to give 4-methyl-6- (1-methylcyclopropoxy) pyridin-3-amine (3.3 g,18.19mmol,74.26% yield, 98.24% purity) as a red oil.
Step 3
At 0 ℃ at N 2 Ac was added in one portion to a mixture of 4-methyl-6- (1-methylcyclopropoxy) pyridin-3-amine (3.3 g,18.52mmol,1 eq.) and Et3N (4.68 g,46.29mmol,6.44mL,2.5 eq.) in DCM (10 mL) 2 O (3.78 g,37.03mmol,3.47mL,2 eq.). The mixture was stirred at 0 ℃ for 30min, then heated to 20 ℃ and stirred for 1 hour. TLC showed the reaction was complete. LCMS showed the desired MS. Saturated NaHCO for reaction 3 Aqueous solution (30 mL) ph=7-8 was adjusted for quenching and with CH 2 Cl 2 (3X 50 mL) extraction. The combined organic phases were washed with brine (3X 50 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 20% -40% ethyl acetate/petroleum ether) to give N- [ 4-methyl-6- (1-methylcyclopropoxy) -3-pyridinyl as a red oil]Acetamide (4 g,18.16mmol,98.08% yield).
Step 4
To N- [ 4-methyl-6- (1-methylcyclopropoxy) -3-pyridinyl at 20 DEG C]To a solution of acetamide (4 g,18.16mmol,1 eq.) in toluene (10 mL) was added KOAc (2.67 g,27.24mmol,1.5 eq.) and Ac 2 O (8.53 g,83.53mmol,7.82mL,4.6 eq.) then the solution was heated to 80℃followed by the dropwise addition of isoamyl nitrite (8.51 g,72.64mmol,9.78mL,4 eq.). The mixture was stirred at 80℃for 2h. TLC and LCMS showed the reaction was complete. The reaction was then filtered with EtOAc (70 mL) and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% ethyl acetate/petroleum ether) to give 1- [5- (1-methylcyclopropoxy) pyrazolo [3,4-c ] as a yellow oil ]Pyridin-1-yl]Ethanone (2.9 g,12.54mmol,69.06% yield).
Step 5
To a solution of 1- [5- (1-methylcyclopropoxy) pyrazolo [3,4-c ] pyridin-1-yl ] ethanone (2.9 g,12.54mmol,1 eq.) in MeOH (50 mL) was added ammonia (7 m,1.79mL,1 eq.) (NH 3 (g)/MeOH) at 20 ℃ in one portion. The mixture was stirred at 20℃for 30min to give a red solution. LCMS showed the reaction was complete. The solution was concentrated in vacuo to give 5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] pyridine (2.1 g, crude) as a yellow solid.
Step 6
To 5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c]To a solution of pyridine (500 mg,2.64mmol,1 eq.) in DMF (30 mL) was added I 2 (1.34 g,5.29mmol,1.06mL,2 eq.) and KOH (444.82 mg,7.93mmol,3 eq.). After addition, the reaction mixture was stirred at 25 ℃ for 16h. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=5:1) showed one major new spot. The reaction mixture was taken up in saturated Na 2 S 2 O 3 (30 mL) was quenched and extracted with ethyl acetate (2X 30 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 15% ethyl acetate/petroleum ether) to give 3-iodo-5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] as a colorless oil ]Pyridine (750 mg,2.38mmol,90.07% yield).
Step 7
At 0 ℃ at N 2 Downward 3-iodo-5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4 ]c]To a mixture of pyridine (750 mg,2.38mmol,1 eq.) in THF (10 mL) was added NaH (114.23 mg,2.86mmol,60% purity, 1.2 eq.) in one portion. The mixture was stirred at 0deg.C for 30min, followed by addition of [ chloro (diphenyl) methyl ]]Benzene (796.22 mg,2.86mmol,1.2 eq.) and the solution was stirred at 20℃for 2h. TLC and LCMS showed the reaction was complete. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -5% (10 min) ethyl acetate/petroleum ether) to give 3-iodo-5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] as a white solid]Pyridine (1.25 g,2.24mmol,94.22% yield).
/>
Step 8
At 20 ℃ at N 2 Downward 3-iodo-5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c]Pyridine (500 mg,896.98umol,1 eq.) Pin 2 B 2 To a mixture of (455.56 mg,1.79mmol,2 eq.) and KOAc (264.09 mg,2.69mmol,3 eq.) in dioxane (5 mL) was added di-tert-butyl (cyclopentyl) phosphorane in one portion; palladium dichloride; iron (29.23 mg,44.85umol,0.05 eq). The mixture was stirred at 80℃for 1 hour. TLC showed the reaction was complete. The mixture was cooled to 25 ℃, filtered and concentrated in vacuo to give 5- (1-methylcyclopropoxy) -3- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1-trityl-pyrazolo [3,4-c ] as a black oil ]Pyridine (800 mg,760.55umol,84.79% yield, 53% purity).
Step 9
To 4- [ [1- (6-chloropyrimidin-4-yl) -4-piperidinyl ] at 25 ℃ under N2]Methyl group]Piperazine-1-carboxylic acid tert-butyl ester (471.57 mg,1.19mmol,1 eq), 5- (1-methylcyclopropoxy) -3- (4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl) -1-trityl-1H-pyrazolo [3,4-c]Pyridine (800 mg,1.19mmol,83% purity, 1 eq.) and di-tert-butyl (cyclopentyl) phosphorane; palladium dichloride; iron (31.05 mg,47.64umol,0.04 eq.) in 1, 4-dioxane (10 mL) and H 2 K was added to the mixture in O (2 mL) at once 3 PO 4 (581.50 mg,2.74mmol,2.3 eq.). The mixture was stirred at 100℃for 16 hours. LCMS showed the desired MS. The mixture was cooled to 25 ℃, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -20% ethyl acetate/petroleum ether) to give 4- ((1- (6- (5- (1-methylcyclopropoxy) -1-trityl-1H-pyrazolo [3, 4-c) as a yellow gum)]Pyridin-3-yl) pyrimidin-4-yl piperidin-4-yl methyl) piperazine-1-carboxylic acid tert-butyl ester (72.5 mg,91.66umol,7.70% yield).
Step 10
To a mixture of tert-butyl 4- [ [1- [6- [5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (72.5 mg,91.66umol,1 eq.) in DCM (5 mL) was added TFA (209.02 mg,1.83mmol,135.73uL,20 eq.) at 20℃in one portion. The mixture was stirred at 20℃for 1h. TLC showed the reaction was complete. The mixture was concentrated in vacuo to give 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -1H-pyrazolo [3,4-c ] pyridine (50 mg,76.43umol,83.39% yield, 86% purity, TFA) as a yellow solid.
Step 11
At 20 ℃ at N 2 Downward 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Pyrimidin-4-yl]-1H-pyrazolo [3,4-c]Pyridine (41 mg,91.40umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl ]]To a mixture of piperidine-4-carbaldehyde (33.76 mg,91.40umol,1 eq.) in MeOH (5 mL) was added borane in one portion; 2-methylpyridine (19.55 mg, 182.81. Mu.l, 2 eq.) and HOAc (5.49 mg, 91.40. Mu.l, 5.23. Mu.l, 1 eq.). The mixture was stirred at 30℃for 1h to give a yellow solution. LCMS showed about 39% of the required MS. The residue was poured into water (5 mL). The aqueous phase was extracted with ethyl acetate (5 ml x 3). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -30%,40 min). After purification by preparative HPLC, the eluate is concentrated and lyophilized to give 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] as a yellow solid]Pyridin-3-yl]Pyrimidin-4-yl]-4-piperidinyl ]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Isoindoline-1, 3-dione (30.3 mg,35.63umol,38.98% yield, 99.70% purity, FA).
Exemplary Synthesis of Compound 179
Step 1
1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-4-yl]Piperidine-4-carbaldehyde (90 mg,253.25umol,1 eq.) and 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl]Piperazin-1-yl]Pyrimidin-4-yl]A mixture of 5- (1-methylcyclopropoxy) -2H-indazole (63.57 mg,126.62umol,0.5 eq. HCl) in HOAC (1 mL) and MeOH (10 mL) was stirred at 20deg.C for 20min, followed by the addition of borane; 2-methylpyridine (54.17 mg,506.49umol,2 eq). The mixture was then brought to 20℃under N 2 Stirred for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Xtime C18 x 30mm x 10um; mobile phase: [ Water (0.225% FA) -ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [4- [4- [ [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (32.5 mg,38.49umol,15.20% yield, 95.34% purity).
Exemplary Synthesis of Compound 180
Step 1
To a solution of 4-bromo-5-fluoro-2-methyl-benzoic acid (1.5 g,6.44mmol,1 eq.) in MeOH (20 mL) was added SOCl 2 (1.91 g,16.09mmol,1.17mL,2.5 eq.). After the addition, the reaction mixture was stirred at 80 ℃ for 1h. LCMS (EB 16-1535-P1 A1) showed the desired MS. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel chromatography (20 g, petroleum ether) to give methyl 4-bromo-5-fluoro-2-methyl-benzoate (1.65 g, crude) as a colorless oil.
Step 2
To a stirred solution of methyl 4-bromo-5-fluoro-2-methyl-benzoate (1.54 g,6.23mmol,1 eq.) in DCE (10 mL) under nitrogen was added NBS (1.33 g,7.48mmol,1.2 eq.) followed by 2- [ (E) - (1-cyano-1-methyl-ethyl) azo]-2-methyl-propionitrile (51.18 mg,311.66umol,0.05 eq) and the resulting mixture was vigorously stirred at 70 ℃ for 2h. LCMS (EB 16-1541-P1 A1) showed no starting material. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: X time C18 x 40mm x 10um; mobile phase: [ Water (0.225% FA) -ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:60% -90%,10 min) to give methyl 4-bromo-2- (bromomethyl) -5-fluoro-benzoate (1.3 g,3.99mmol,63.98% yield) as a white solid.
Step 3
At 20 ℃ at N 2 To a mixture of methyl 4-bromo-2- (bromomethyl) -5-fluoro-benzoate (1.3 g,3.99mmol,1 eq.) and 3-aminopiperidine-2, 6-dione (787.71 mg,4.79mmol,1.2 eq., HCl) in DMF (10 mL) was added DIEA (2.58 g,19.94mmol,3.47mL,5 eq.) in one portion. The mixture was stirred at 85 ℃ for 48 hours. LCMS (EB 16-1548-P1 A2) showed the desired MS. The crude product was concentrated in vacuo. The crude product was purified by MeCN (20 mL) and H 2 O (20 mL) was ground at 20deg.C. The crude was filtered and the solid concentrated in vacuo to give 3- (5-bromo-6-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (830 mg,2.43mmol,61.01% yield) as a dark grey solid.
Step 4
To a solution of 3- (5-bromo-6-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (100 mg,293.14umol,1 eq) and 4- (dimethoxymethyl) piperidine (93.35 mg,586.28umol,2 eq) in DMSO (5 mL) was added Cs 2 CO 3 (191.02 mg,586.28umol,2 equivalents) and Pd-PEPSI-pent Cl-o-methylpyridine (15.92 mg,29.31umol,0.1 equivalents). After addition, the reaction mixture was taken up in N at 80 ℃ 2 Stirring was carried out for 12h. LCMS (EB 16-1555-P1 A2) showed the desired MS. The mixture was cooled to 20 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Drying, filtering andconcentrated in vacuo. The residue was purified by preparative TLC (petroleum ether/ethyl acetate=0/1) to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl as a yellow oil]-6-fluoro-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (70 mg,91.79umol,31.31% yield, 55% purity).
Step 5
At 20 ℃ at N 2 Downward 3- [5- [4- (dimethoxymethyl) -1-piperidinyl]-6-fluoro-1-oxo-isoindolin-2-yl]To a mixture of piperidine-2, 6-dione (70 mg, 166.89. Mu.L, 1 eq.) in THF (5 mL) was added HCl (2M, 83.44. Mu.L, 1 eq.) in one portion. The mixture was stirred at 20 ℃ for 2 hours to give a yellow solution. TLC showed the reaction was complete. Pouring the residue into saturated NaHCO 3 To adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 1- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl as a yellow solid ]Piperidine-4-carbaldehyde (43 mg,63.34umol,37.95% yield, 55% purity).
Step 6
At 20 ℃ at N 2 Downward 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Pyrimidin-4-yl]-2H-indazole (50 mg,111.71umol,0.97 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl]To a mixture of piperidine-4-carbaldehyde (43 mg,115.16umol,1 eq.) in MeOH (10 mL) was added borane in one portion; 2-methylpyridine (24.64 mg, 230.33. Mu.mol, 2 eq.) and HOAc (6.92 mg, 115.16. Mu.mol, 6.59. Mu.L, 1 eq.). The mixture was stirred at 20℃for 10h to give a yellow solution. LCMS (EB 16-1563-P1)A1 A desired MS is shown. Pouring the obtained product into H 2 O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by preparative HPLC (column: phenomenex luna 30*30mm*10um+YMC AQ 100*30*10um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:2% -42%,26 min) to give 3- [ 6-fluoro-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]-4-piperidinyl ]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (12.4 mg,15.32umol,13.31% yield, 99.47% purity).
Exemplary Synthesis of Compound 181
Step 1
To a solution of 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (1 g,2.32mmol,1.32 eq.) and tert-butyl 4- (4-piperidinylmethyl) piperazine-1-carboxylate (500 mg,1.76mmol,1 eq.) in DMSO (8 mL) was added DIEA (684.05 mg,5.29mmol,921.90ul,3 eq.). After the addition, the reaction solution was stirred at 90 ℃ for 1h. LCMS (EB 12-979-P1B) and TLC (petroleum ether: ethyl acetate=1:1) showed that the reaction was complete. After cooling, the reaction mixture was diluted with ethyl acetate (20 mL) and washed with brine (20 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 68% ethyl acetate/petroleum ether) to give tert-butyl 4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (660 mg,876.17umol,49.66% yield, 90% purity) as a white solid.
Step 2
To a solution of tert-butyl 4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (658.04 mg,970.64umol,1 eq.) in DCM (5 mL) was added HCl/EtOAc (4M, 5 mL). After the addition, the reaction solution was stirred at 20 ℃ for 12h. LCMS (EB 12-983-P1B) showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -1H-indazole (450 mg, crude, HCl) as a white solid. The crude product was used directly in the next step.
Step 3
At 0 ℃ at N 2 To a solution of TMP (4.84 g,34.25mmol,5.81mL,2.5 eq.) in THF (30 mL) was added n-BuLi (2.5M, 13.70mL,2.5 eq.) dropwise and the resulting mixture stirred at 0deg.C for 30min. The reaction mixture was then cooled to-45 ℃, 4-bromo-3-fluoro-benzoic acid (3 g,13.70mmol,1 eq.) dissolved in THF (15 mL) was added dropwise and stirring was continued for 5h at-45 ℃. DMF (1.50 g,20.55mmol,1.58mL,1.5 eq.) was added dropwise and the reaction mixture was warmed to 20℃and stirred for 12h. LCMS (EB 12-978-P1B 1) showed consumption of starting material. The reaction mixture was quenched by 3M HCl (25 mL) at 0 ℃ and extracted with ethyl acetate (30 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 58% ethyl acetate/petroleum ether) to give 5-bromo-4-fluoro-3-hydroxy-3H-isobenzofuran-1-one (1.7 g,6.88mmol,50.24% yield) as a brown solid.
Step 4
To a solution of 5-bromo-4-fluoro-3-hydroxy-3H-isobenzofuran-1-one (500 mg,2.02mmol,1 eq.) in DMF (5 mL) was added 3-aminopiperidine-2, 6-dione hydrochloride (499.73 mg,3.04mmol,1.5 eq.) and NaBH (OAc) 3 (1.07 g,5.06mmol,2.5 eq.). After addition, the reaction mixture was stirred at 20 ℃ for 12h. LCMS (EB 12-984-P1B) showed the reaction was complete. The reaction mixture was diluted with water (30 mL) and cooled to 0 ℃ with a water/ice bath, which resulted in the formation of a precipitate. The resulting mixture was filtered and the dark blue solid was washed with MTBE (30 ml x 2). The obtained solid was dried under reduced pressure to give 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (340 mg, crude) as a blue solid. The crude product was used directly in the next step.
Step 5
To a solution of 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (200 mg,586.28umol,1 eq.) and 4- (dimethoxymethyl) piperidine (186.70 mg,1.17mmol,2 eq.) in DMSO (5 mL) was added Cs 2 CO 3 (382.05 mg,1.17mmol,2 eq.) and Pd-PEPSI-pent Cl-o-methylpyridine (31.84 mg,58.63umol,0.1 eq.). After addition, the reaction mixture was taken up in N at 80 ℃ 2 Stirring was carried out for 12h. LCMS (EB 12-992-P1B) and TLC (petroleum ether: ethyl acetate=0:1) showed that the reaction was complete. After cooling, the reaction mixture was poured into saturated NH 4 Cl (40 mL) and extracted with ethyl acetate (20 mL. Times.3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative TLC (pure ethyl acetate) to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl as a yellow solid]-4-fluoro-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (50 mg,119.20umol,20.33% yield).
Step 6
To 3- [5- [4- (dimethoxymethyl) -1-piperidinyl]-4-fluoro-1-oxo-isoindolin-2-yl]To a solution of piperidine-2, 6-dione (50 mg,119.20umol,1 eq.) in THF (2 mL) was added HCl (2M, 2mL,33.56 eq.). After the addition, the reaction solution was stirred at 20 ℃ for 2h. LCMS showed the reaction was complete. The reaction mixture was passed through saturated NaHCO 3 (10 mL) quenched and extracted with ethyl acetate (10 mL. Times.3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure to give 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl as a yellow solid]Piperidine-4-carbaldehyde (60 mg, crude). The crude product was used directly in the next step.
Step 7
To a mixture of 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (15 mg,40.17 mol,1.30 eq.) in MeOH (5 mL) and HOAc (0.5 mL) was added a solution of 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -1H-indazole (15 mg,30.99 mol,1 eq., HCl) and DIEA (4.01 mg,30.99 mol,5.40ul,1 eq.) in MeOH (1 mL). Then borane is added; 2-methylpyridine (6.63 mg, 61.98. Mu. Mol,2 eq.) was added to the above solution. After the addition, the reaction solution was stirred at 20 ℃ for 12h. LCMS showed the reaction was complete. The reaction solution was concentrated under reduced pressure to remove most of the solvent. The residue was purified by preparative HPLC (column: xtime C18. Times.30 mm. Times.10 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 3- [ 4-fluoro-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (6.9 mg,8.51 mol,27.46% yield, 99.29% purity) as a white solid.
Exemplary Synthesis of Compound 182
Step 1
To a mixture of 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (45 mg,120.52umol,1.34 eq.) in MeOH (5 mL) and HOAc (0.5 mL) was added a solution of 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (45 mg,89.64umol,1 eq., HCl) and DIEA (11.58 mg,89.64umol,15.61ul,1 eq.) in MeOH (1 mL). Then borane is added; 2-methylpyridine (19.18 mg,179.27umol,2 eq.) was added to the above solution. After the addition, the reaction solution was stirred at 20 ℃ for 12h. LCMS showed the reaction was complete. The reaction solution was concentrated under reduced pressure to remove most of the solvent. The residue was purified by preparative HPLC (column: phenomenex luna 30*30mm*10um+YMC AQ 100*30*10um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -40%,26 min) to give 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (17.0 mg,20.53umol,22.90% yield, 99.38% purity) as a white solid.
Exemplary Synthesis of Compound 183
Step 1:
at 20 ℃ at N 2 Downward 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ]]Piperazin-1-yl]Pyrimidin-4-yl]-5- (1-methylcyclopropoxy) -2H-indazole (87.28 mg,187.48umol,1 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl]To a mixture of piperidine-4-carbaldehyde (70 mg,187.48umol,1 eq.) in MeOH (10 mL) was added borane in one portion; 2-methylpyridine (20).05mg, 187.48. Mu.l, 1 eq.) and HOAc (11.26 mg, 187.48. Mu.l, 10.72. Mu.l, 1 eq.). The mixture was stirred at 20 ℃ for 10 hours to give a yellow solution. LCMS showed the desired mass. The reaction solution was concentrated under reduced pressure to remove most of the solvent. The residue was purified by preparative HPLC (column: xtime C18100X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 6-fluoro-5- [4- [ [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (30.7 mg,35.72umol,19.05% yield, 95.75% purity).
Exemplary Synthesis of Compound 184
Step 1:
to a solution of 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (240 mg,703.54umol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester (196.55 mg,1.06mmol,1.5 eq.) in DMSO (10 mL) was added Cs 2 CO 3 (458.45 mg,1.41mmol,2 eq.) and Pd-PEPSI-pent Cl-o-methylpyridine (38.20 mg,70.35umol,0.1 eq.) the mixture was then stirred at 80℃for 16 hours. TLC (petroleum ether: ethyl acetate=1:3, r f =0.2, uv=254 nm) shows that new spots formed. The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layers were washed with brine (50 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel12g/>Silica gel flash column, eluent: gradient of 0% -60% ethyl acetate/petroleum ether, 40 mL/min)And (5) purifying. Obtaining the compound 4- [2- (2, 6-dioxo-3-piperidyl) -4-fluoro-1-oxo-isoindolin-5-yl) as a pale yellow solid]Piperazine-1-carboxylic acid tert-butyl ester (90 mg,133.04umol,18.91% yield, 66% purity). />
Step 2:
to a solution of 4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperazine-1-carboxylic acid tert-butyl ester (260 mg,582.34umol,1 eq.) in TFA (66.40 mg,582.34umol,43.12uL,1 eq.) the mixture was stirred at 25℃for 2 hours. LCMS showed the desired compound and the starting material was completely consumed. The reaction mixture was concentrated under reduced pressure to give a residue. The compound 3- (4-fluoro-1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (300 mg,358.40umol,61.54% yield, 55% purity, TFA) was obtained as a pale yellow oil.
Step 3:
to a solution of benzyl (3S) -3-methylpiperazine-1-carboxylate (1.6 g,6.83mmol,1 eq.) and 2-chloroacetaldehyde (6.70 g,34.15mmol,5.49mL,40% purity, 5 eq.) in DCM (15 mL) and MeOH (15 mL) was added HOAc (41.01 mg,682.90umol,39.06uL,0.1 eq.) followed by stirring the mixture at 25℃for 0.5 h followed by NaBH 3 CN (858.30 mg,13.66mmol,2 eq.) was added to the above mixture and the mixture was stirred at 25℃for 0.5 h. TLC (petroleum ether: ethyl acetate=1:1, rf=0.52, i 2 ) The main spot is shown to be formed. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel40g />Silica gel flash column, eluent: 0% -44% ethyl acetate/petroleum ether gradient, 50 mL/min). The compound (3S) -4- (2-chloroethyl) -3-methyl-piperazine-1-carboxylic acid benzyl ester (1.8 g,6.06mmol,88.81% yield) was obtained as a colorless oil.
Step 4:
benzyl (3S) -4- (2-chloroethyl) -3-methyl-piperazine-1-carboxylate (800 mg,2.70mmol,1 eq.) and 4- (dimethoxymethyl) piperidine (515.03 mg,3.23mmol,1.2 eq.) were dissolved in EtOH (10 mL) and H 2 O (1 mL), then NaHCO 3 (679.32 mg,8.09mmol,314.50uL,3 eq.) was added to the reaction and stirred at 80℃for 5 hours. TLC (dichloromethane: methanol=5:1, rf=0.52, pma) showed that the main spot formed and the starting material was completely consumed. To the reaction mixture was added water (50 mL) and extracted with brine (50 mL x 3). The combined organic layers were washed with brine (80 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel12g/>Silica gel flash column, eluent: 0% -12% ethyl acetate/petroleum ether gradient, 50 mL/min). The compound (3S) -4- [2- [4- (dimethoxymethyl) -1-piperidinyl was obtained as a colorless oil]Ethyl group]-3-methyl-piperazine-1-carboxylic acid benzyl ester (0.4 g,953.39umol,35.37% yield). />
Step 5:
to (3S) -4- [2- [4- (dimethoxymethyl) -1-piperidinyl]Ethyl group]To a solution of benzyl 3-methyl-piperazine-1-carboxylate (400 mg,953.39umol,1 eq.) in THF (5 mL) was added HCl (2 m,5mL,10.49 eq.) and the mixture was stirred at 25 ℃ for 2 hours. The reaction mixture was taken up with saturated NaHCO 3 Dilute to ph=8 and extract with EtOAc (50 ml x 3). The combined organic layers were washed with brine (50 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The compound (3S) -4- [2- (4-formyl-1-piperidinyl) ethyl was obtained as a pale yellow oil]-3-methyl-piperazine-1-carboxylic acid benzyl ester (350 mg,937.11umol,98.29% yield).
Step 6:
to 3- (4-fluoro-1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (300 mg,651.64umol,1 eq, TFA) and (3S) -4- [2- (4-formyl-1-piperidinyl) ethyl ]To a solution of benzyl 3-methyl-piperazine-1-carboxylate (243.38 mg,651.64umol,1 eq.) in DCM (5 mL) and MeOH (1 mL) was added NaOAc (267.28 mg,3.26mmol,5 eq.) followed by stirring the mixture for 1 hour and adding NaBH (OAc) to the solution 3 (276.22 mg,1.30mmol,2 eq.) the mixture was then stirred at 25℃for 2 hours. LCMS showed about 54% of the desired compound and TLC (dichloromethane: methanol=5:1, r f =0.28,I 2 ) The main spot is shown to be formed. The reaction mixture was taken up with saturated NaHCO 3 Dilute to ph=8 and extract with EtOAc (50 ml x 3). The combined organic layers were washed with brine (50 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel12g />Silica gel flash column, eluent: 0% -14% methylene chloride: methanol ether gradient, 40 mL/min). Obtaining the compound (3S) -4- [2- [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a white solid]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Ethyl group]-3-methyl-piperazine-1-carboxylic acid benzyl ester (150 mg,213.12umol,32.70% yield). />
Step 7:
to a solution of (3S) -4- [2- [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] ethyl ] -3-methyl-piperazine-1-carboxylic acid benzyl ester (140 mg,198.91 mol,1 eq) in TFA (4.62 g,40.52mmol,3ml,203.70 eq) was then stirred at 65 ℃ for 0.5 h. LCMS showed about 87.2% of the desired compound and the starting material was completely consumed. The reaction mixture was concentrated under reduced pressure to give a residue. The compound 3- [ 4-fluoro-5- [4- [ [1- [2- [ (2S) -2-methylpiperazin-1-yl ] ethyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (140 mg,186.94umol,93.99% yield, 91.3% purity, TFA) was obtained as a pale yellow oil.
Step 8:
to a solution of 3- [ 4-fluoro-5- [4- [ [1- [2- [ (2S) -2-methylpiperazin-1-yl ] ethyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (140 mg,204.76umol,1 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (61.58 mg,204.76umol,1 eq) in DMSO (3 mL) was added DIEA (132.32 mg,1.02mmol,178.33ul,5 eq), and the mixture was stirred at 90 ℃ for 16 hours. LCMS showed about 40.8% of the desired compound and the starting material was completely consumed. The residue was purified by preparative HPLC (column: xtimate C18 x 30mm x 10um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -25%,40 min). The compound 3- [ 4-fluoro-5- [4- [ [1- [2- [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] ethyl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] -1-oxo-isoindol-2-yl ] piperidine-2, 6-dione (31.3 mg,37.40umol,18.26% yield, 99.65% purity) was obtained as a brown solid.
Exemplary Synthesis of Compound 185
Step 1:
to 1- [2- (2, 6-dioxo-3-piperidyl) -6-fluoro-1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (80 mg,156.41umol,73% purity, 1 eq.) to a mixture of MeOH (5 mL) and HOAc (0.5 mL) was added 6-fluoro-5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ]Pyrimidin-4-yl]A solution of 1H-indazole (78.52 mg, 156.41. Mu.mol, 1 eq. HCl) and DIEA (20.21 mg, 156.41. Mu.L, 27.24. Mu.L, 1 eq.) in MeOH (0.5 mL). Then borane is added; 2-methylpyridine (33.46 mg,312.82umol,2 eq.) was added to the above solution. After the addition, the reaction solution was stirred at 40 ℃ for 12 hours. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove most of the solvent. The residue was purified by preparative HPLC (column: xtime C18 x 30mM x 10um; mobile phase: [ water (10 mM NH 4 HCO 3 )-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:20% -70%,40 min) to afford 3- [ 6-fluoro-5- [4- [ [4- [ [1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as an off-white solid]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (50.4 mg,61.24umol,39.16% yield, 100% purity).
Exemplary Synthesis of Compound 186
Step 1:
to 6-fluoro-5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Pyrimidin-4-yl]-2H-indazole (80.67 mg,160.70umol,1 eq, HCl) 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindol-5-yl was added to a mixture of HOAC (1 mL) and MeOH (10 mL) ]A solution of piperidine-4-carbaldehyde (60 mg, 160.70. Mu. Mol,1 eq.) and DIEA (62.31 mg, 482.09. Mu. Mol, 83.97. Mu.L, 3 eq.) in MeOH (0.5 mL). Then borane is added; 2-methylpyridine (34.38 mg,321.39umol,2 eq.) was added to the above solution. After the addition, the reaction solution was stirred at 20 ℃ for 16 hours. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: xtime C18 x 30mM x 10um; mobile phase: [ water (10 mM NH 4 HCO 3 )-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:30% -80%,40 min) to give 3- [ 4-fluoro-5- [4- [ [4- [ [1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (54.2 mg,63.52umol,39.53% yield, 96.44% purity).
Exemplary Synthesis of Compound 187
Step 1:
to 3- [6- [ (3S) -4- [ (4-fluoro-4-piperidinyl) methyl]-3-methyl-piperazin-1-yl]Pyrimidin-4-yl]-5- (1-methylcyclopropoxy) -1H-indazole (64 mg,133.45umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (63.07 mg,133.45umol,79% purity, 1 eq.) was added borane to a mixture of MeOH (10 mL), acOH (1 mL) and THF (10 mL); 2-methylpyridine (28.55 mg,266.89umol,2 eq) . The mixture was stirred at 20℃for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18100X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (16.6 mg,19.21umol,14.40% yield, 96.878% purity).
Exemplary Synthesis of Compound 188
Step 1:
to a solution of 3- (5-bromo-6-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (400 mg,1.17mmol,1 eq.) and tert-butyl 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carboxylate (530.12 mg,1.76mmol,1.5 eq.) in DMSO (10 mL) was added Cs 2 CO 3 (764.09 mg,2.35mmol,2 eq.) and Pd-PEPSI-pent Cl-o-methylpyridine (100.91 mg,117.26umol,0.1 eq.). After addition, the reaction mixture was taken up in N at 80 ℃ 2 Stirred for 12 hours. LCMS and TLC showed the reaction was complete. After cooling, the reaction mixture was filtered, and the filtrate was diluted with ethyl acetate (50 mL). The organic layer was washed with brine (30 ml x 2), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give 4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] as a grey solid]Piperazin-1-yl]Methyl group]-4-fluoro-piperidine-1-carboxylic acid tert-butyl ester (240 mg,393.15umol,33.53% yield, 92% purity).
Step 2:
to a solution of 4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -4-fluoro-piperidine-1-carboxylic acid tert-butyl ester (240 mg,427.34umol,1 eq.) in DCM (3 mL) was added HCl/EtOAc (4M, 4mL,37.44 eq.). After addition, the reaction was stirred at 20 ℃ for 2 hours. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 3- [ 6-fluoro-5- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (220 mg,384.36umol,89.94% yield, 87% purity, HCl) as a grey solid. The crude product was used directly in the next step.
Step 3:
to a mixture of 3- [ 6-fluoro-5- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (90 mg,157.24umol,87% purity, 1 eq, HCl) in MeOH (10 mL) and HOAc (1 mL) was added a solution of 1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (62 mg,164.27umol,1.04 eq) and DIEA (20.32 mg,157.24umol,27.39ul,1 eq) in MeOH (1 mL). Then borane is added; 2-methylpyridine (33.64 mg,314.48umol,2 eq.) was added to the above solution. After the addition, the reaction solution was stirred at 25 ℃ for 12 hours. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove most of the solvent. The residue was purified by preparative HPLC (column: xtime C18. Times.30 mm. Times.10 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,40 min) to afford 3- [ 6-fluoro-5- [4- [ [ 4-fluoro-1- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (37.3 mg,45.33umol,28.83% yield, 100% purity) as an off-white solid.
Exemplary Synthesis of Compound 189
Step 1:
to 2- [ [3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl]Methoxy group]To a mixture of ethyl-trimethyl-silane (200 mg,445.44umol,1 eq) and tert-butyl 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carboxylate (134.26 mg,445.44umol,1 eq) in DMSO (3 mL) was added TEA (135.22 mg,1.34mmol,186.00uL,3 eq). The mixture was stirred at 90℃for 16 hours. LCMS showed the desired MS. The residue was poured into water (30 mL). The aqueous phase was extracted with ethyl acetate (30 ml x 3). The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -28% (15 min) ethyl acetate/petroleum ether, 0% -28% (10 min) ethyl acetate/petroleum ether) to give 4-fluoro-4- [ [4- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] as a yellow gum]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]Tert-butyl piperidine-1-carboxylate (250 mg,226.87umol,50.93% yield, 64.789% purity).
Step 2:
to a mixture of tert-butyl 4-fluoro-4- [ [4- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (250 mg,350.17umol,1 eq.) in EtOAc (2 mL) was added HCl/EtOAc (4 m,87.54ul,1 eq.). The mixture was stirred at 40℃for 2 hours. TLC showed the reaction was complete. The residue was filtered and concentrated in vacuo to give 6-fluoro-3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (200 mg, crude, HCl) as a white solid.
Step 3:
to 6-fluoro-3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl]Piperazin-1-yl]Pyrimidin-4-yl]-5- (1-methylcyclopropoxy) -2H-indazole (85 mg,175.78umol,1.38 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (60 mg,126.95umol,79% purity, 1 eq.) was added borane to a mixture of MeOH (10 mL), THF (10 mL) and AcOH (1 mL); 2-methylpyridine (27.16 mg,253.90umol,2 eq). The mixture was stirred at 40℃for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-4- [ [4- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazol-3-yl) as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (55.4 mg,65.83umol,51.86% yield, 99.927% purity).
Exemplary Synthesis of Compound 190
Step 1:
3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (200 mg,586.28umol,1 eq.) and 4-fluoro-4- (piperazin-1-ylmethyl) piperidine-1-carboxylic acid tert-butyl ester (176).71mg,586.28umol,1 eq.) Cs 2 CO 3 A mixture of (382.04 mg,1.17mmol,2 eq.) and Pd-PEPSI-pent Cl-o-methylpyridine (25.23 mg,29.31umol,0.05 eq.) in DMSO (3 mL) was degassed and purged 3 times with N2, then the mixture was N at 80 ℃ 2 Stirring is carried out for 16 hours under an atmosphere. LC-MS (EB 2049-281-P1B) showed complete consumption of reactant 1. After cooling, the reaction mixture was filtered, and the filtrate was diluted with THF (5 mL) and washed with brine (5 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure.
The residue was purified by flash chromatography on silica gel12g/>Silica gel flash column, eluent: 0% -35% ethyl acetate/petroleum ether gradient, 75 mL/min) to afford 4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a white solid]Piperazin-1-yl]Methyl group]-4-fluoro-piperidine-1-carboxylic acid tert-butyl ester (100 mg,178.06umol,30.37% yield).
Step 2:
to a solution of 4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -4-fluoro-piperidine-1-carboxylic acid tert-butyl ester (95 mg,169.15umol,1 eq.) in DCM (3 mL) was added TFA (57.86 mg,507.46umol,37.57uL,3 eq.). The mixture was stirred at 25℃for 1 hour. TLC indicated no residue of reactant 1 and a major new spot of greater polarity was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent. No further purification was necessary. 3- [ 4-fluoro-5- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (83 mg,87.97umol,52.01% yield, 61% purity, TFA) was obtained as a white solid.
Step 3:
to 1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl]Pyrimidin-4-yl]Piperidine-4-carbaldehyde (33 mg,87.43umol,1 eq.) and 3- [ 4-fluoro-5- [4- [ (4-fluoro-4-piperidinyl) methyl]Piperazin-1-yl]-1-oxo-isoindolin-2-yl]To a solution of piperidine-2, 6-dione (50.32 mg,87.43umol,1 eq, TFA) in AcOH (0.5 mL) and MeOH (3 mL) was added borane; 2-methylpyridine (18.70 mg,174.86umol,2 eq). The mixture was stirred at 25℃for 16 hours. LC-MS (EB 2049-284-P1A) showed complete consumption of reactant 1 and the required mass was detected. The mixture was poured onto ice water (5 mL) and extracted with EtOAc (5 ml×2). The organic layer was washed with brine (5 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The crude product was purified by reverse phase HPLC (column: xtimate C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -30%,40 min) to give 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-1- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl) as a white solid]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]-4-piperidinyl]Methyl group]Piperazin-1-yl]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (42.4 mg,51.33umol,58.70% yield, 99.619% purity).
Exemplary Synthesis of Compound 191
Step 1:
to a solution of 3-iodo-5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine (2 g,3.59mmol,1 eq.) in toluene (20 mL) under nitrogen was added hexamethylditin (2.35 g,7.18mmol,1.5mL,2 eq.) and 4-di-tert-butylphosphoranyl-N, N-dimethyl-aniline; palladium dichloride (254 mg,0.36mmol,0.25mL,0.1 eq.). The reaction was stirred at 140 ℃ under microwaves for 2 hours. TLC (petroleum ether: ethyl acetate=5:1) showed that a new spot was detected. The mixture was diluted with ethyl acetate (100 mL) and filtered through celite. The filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=1:0 to 10:1). Trimethyl- [5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridin-3-yl ] stannane (1.8 g,3.03mmol,84% yield) was obtained as a pale yellow oil.
Step 2:
to a solution of trimethyl- [5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridin-3-yl ] stannane (300 mg,0.5mmol,1 eq.) and 4, 6-dichloropyrimidine (113 mg,0.76mmol,1.5 eq.) in toluene (3 mL) under nitrogen was added thiophene-2-carbonyloxy copper (192 mg,1.01mmol,2 eq.) and 1, 1-bis (diphenylphosphino) ferrocene-palladium (II) dichloride dichloromethane complex (41.mg, 0.05mmol,0.1 eq.). The reaction was stirred at 140 ℃ under microwaves for 15 minutes. LCMS showed detection of the desired MS. The mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=1:0 to 5:1). 3- (6-Chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine (140 mg,0.26mmol,51% yield) was obtained as a pale yellow solid.
Step 3:
to a solution of 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine (120 mg,0.22mmol,1 eq.) and diisopropylethylamine (85 mg,0.66mmol,0.1mL,3 eq.) in dimethyl sulfoxide (3 mL) was added 4- (dimethoxymethyl) piperidine (42 mg,0.26mmol,1.2 eq.). The reaction mixture was stirred at 120℃for 6 hours. LCMS showed detection of the desired MS. The mixture was poured into water (20 mL) and the aqueous phase extracted with ethyl acetate (20 mL x 3). The combined organic phases were washed with brine (20 ml x 2), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by preparative TLC (petroleum ether: ethyl acetate=2:1). 3- [6- [4- (dimethoxymethyl) -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine (95 mg,0.14mmol,64% yield) was obtained as a pale yellow solid.
Step 4:
to a solution of 3- [6- [4- (dimethoxymethyl) -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine (110 mg,0.16mmol,1 eq.) in tetrahydrofuran (3 mL) was added a sulfuric acid solution (3 m,1.2mL,22.23 eq.). The reaction was stirred at 40℃for 7 hours. LCMS showed detection of the desired MS. Saturated sodium bicarbonate (50 mL) was added to the mixture and the aqueous phase was extracted with ethyl acetate (50 mL x 3). The combined organic phases were washed with brine (50 mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was used directly in the next step. 1- [6- [5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (60 mg, crude) was obtained as a pale yellow solid.
Step 5:
to a solution of 4-bromo-5-fluoro-2-methyl-benzoic acid (2 g,8.58mmol,1 eq.) in methanol (20 mL) was added sulfuric acid solution (426 mg,4.29mmol,0.2mL,0.5 eq.). The reaction was stirred at 80℃for 12 hours. TLC (petroleum ether: ethyl acetate=3:1) showed that a new spot was detected. The mixture was poured into saturated sodium bicarbonate (50 mL) and the aqueous phase was extracted with ethyl acetate (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=1:0 to 20:1). 4-bromo-5-fluoro-2-methyl-benzoic acid methyl ester (2.3 g, crude product) was obtained as a colorless oil.
Step 6:
to a solution of methyl 4-bromo-5-fluoro-2-methyl-benzoate (2.3 g,9.31mmol,1 eq.) in carbon tetrachloride (25 mL) was added N-bromosuccinimide (1.66 g,9.31mmol,1 eq.) and 2, 2-azobisisobutyronitrile (153 mg,0.93mmol,0.1 eq.). The reaction mixture was stirred at 80℃for 12 hours. TLC (petroleum ether: ethyl acetate=5:1) showed that a new spot was detected. The mixture was filtered and the filter cake was washed with ethyl acetate (20 ml x 2). The filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=1:0 to 20:1). 4-bromo-2- (bromomethyl) -5-fluoro-benzoic acid methyl ester (1.9 g,5.83mmol,62% yield) was obtained as a white solid.
Step 7:
to a solution of methyl 4-bromo-2- (bromomethyl) -5-fluoro-benzoate (1.9 g,5.83mmol,1 eq.) and N, N-diisopropylethylamine (3.77 g,29.14mmol,5.08mL,5 eq.) in N, N-dimethylformamide (30 mL) was added 3-aminopiperidine-2, 6-dione; hydrochloride (1.01 g,6.12mmol,1.05 eq.). The reaction mixture was stirred at 40 ℃ for 1 hour and at 100 ℃ for 11 hours. LCMS showed detection of the desired MS. The mixture was poured into water (150 mL), the mixture was filtered, and the filter cake was dried in vacuo. The mixture was triturated with petroleum ether and ethyl acetate (V/v=1:1, 20 mL). 3- (5-bromo-6-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (1.25 g,3.45mmol,59% yield, 94% purity) was obtained as a grey solid.
Step 8:
to a solution of 3- (5-bromo-6-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (300 mg,0.88mmol,1 eq), tert-butyl 4- (4-piperidinylmethyl) piperazine-1-carboxylate (299 mg,1.06mmol,1.2 eq) in dimethyl sulfoxide (6 mL) was added cesium carbonate (573 mg,1.76mmol,2 eq) and Pd-PEPPSI-pentcl-o-methylpyridine (85 mg,0.09mmol,0.1 eq) under nitrogen. The reaction was stirred at 100℃for 12 hours. LCMS showed detection of the desired MS. Dichloromethane (20 mL) and water (20 mL) were added to the mixture and the aqueous phase was extracted with dichloromethane (20 mL x 2). The combined organic phases were washed with brine (20 mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by preparative TLC (dichloromethane: methanol=10:1). 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylic acid tert-butyl ester (120 mg,0.20mmol,23% yield, 93% purity) was obtained as a grey solid.
Step 9:
to a solution of tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (60 mg,0.11mmol,1 eq.) in dichloromethane (1 mL) was added a solution of hydrochloric acid (4M in dioxane, 2.4mL,86.98 eq.). The reaction was stirred at 20℃for 1 hour. TLC (dichloromethane: methanol=10:1) showed that a new spot was detected. The mixture was concentrated in vacuo. The residue was used directly in the next step. 3- [ 6-fluoro-1-oxo-5- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindolin-2-yl ] piperidine-2, 6-dione (52 mg,0.11mmol,98% yield, hydrochloride) was obtained as a grey solid.
Step 10:
to a solution of 3- [ 6-fluoro-1-oxo-5- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindol-2-yl ] piperidine-2, 6-dione (51 mg,0.10mmol,1 eq, hydrochloride) in methanol (3 mL) was added sodium acetate (43 mg,0.53mmol,5 eq), followed by 1- [6- [5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (40 mg,0.10mmol,1 eq) and acetic acid (0.3 mL). The mixture was then stirred at 20℃for 0.5 h. Boranes; 2-methylpyridine (22 mg,0.21mmol,2 eq.) was added to the mixture and the reaction mixture was stirred at 20℃for 2 hours. LCMS showed detection of the desired MS. Ethyl acetate (30 mL) was added to dilute the mixture, and water (20 mL) was added to the mixture. The aqueous phase was extracted with ethyl acetate (30 ml x 2). The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: unisil3-100C18 Ultra 150*50mm*3um; mobile phase: [ water (0.225% FA) -ACN ]; B%:6% -36%,10 min). 3- [ 6-fluoro-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (31.1 mg,0.04mmol,34.19% yield, 99% purity, formate) was obtained as a white solid.
Exemplary Synthesis of Compound 192
Step 1:
the 6-fluoro-3- [6- [ (3S) -4- [ (4-fluoro-4-piperidinyl) methyl]-3-methyl-piperazin-1-yl]Pyrimidin-4-yl]-5- (1-methylcyclopropoxy) -2H-indazole (72.64 mg,102.19umol,70% purity, 1.07 eq.) in weight) And 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl]A mixture of piperidine-4-carbaldehyde (45 mg,95.21umol,79% purity, 1 eq.) in HOAC (1 mL) and MeOH (10 mL) was stirred at 20deg.C for 20 min, followed by the addition of borane; 2-methylpyridine (20.37 mg, 190.42. Mu. Mol,2 eq.). The mixture was then brought to 20℃under N 2 Stirred for 16 hours. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-4- [ [ (2S) -4- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazol-3-yl as a white solid]Pyrimidin-4-yl]-2-methyl-piperazin-1-yl]Methyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (67.6 mg,77.60umol,81.50% yield, 98.14% purity).
Exemplary Synthesis of Compound 193
Step 1:
to 2- [ [3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl]Methoxy group]To a mixture of ethyl-trimethyl-silane (200 mg,445.44umol,1 eq), 4- (dimethoxymethyl) piperidine (85.11 mg,534.53umol,1.2 eq) in DMSO (5 mL) was added DIEA (57.57 mg,445.44umol,77.59uL,1 eq) followed by subjecting the mixture to N at 100deg.C 2 Stirring is carried out for 12 hours under an atmosphere. LCMS showed the desired mass. TLC (petroleum ether: ethyl acetate=3:1) showed a new major spot. Pouring the obtained product into H 2 O (10 mL). The mixture was extracted with ethyl acetate (50 ml x 3). The organic phase was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by silica gel column chromatography (0% to 27% ethyl acetate/petroleum ether) to give 2- [ [3- [6- [4- (dimethoxymethyl) -1-piperidinyl ] as a yellow oil]Pyrimidin-4-yl]-6-fluoro-5- (1-methyl ring)Propoxy) indazol-2-yl]Methoxy group]Ethyl-trimethyl-silane (210 mg,367.29umol,82.45% yield).
Step 2:
to 2- [ [3- [6- [4- (dimethoxymethyl) -1-piperidinyl]Pyrimidin-4-yl]-6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl ]Methoxy group]To a solution of ethyl-trimethyl-silane (210 mg,367.29umol,1 eq.) in THF (10 mL) was added HCl (2 m,2mL,10.89 eq.). After the addition, the reaction solution was stirred at 60 ℃ for 2 hours. LCMS showed the desired MS. After cooling, the residue was poured into NaHCO 3 To adjust ph=7-8, and washed with water (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (20 mL), dried over anhydrous Na 2 SO 4 Drying and concentration in vacuo afforded 1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -2H-indazol-3-yl as a colorless oil]Pyrimidin-4-yl]Piperidine-4-carbaldehyde (140 mg,343.42umol,93.50% yield, 97% purity).
Step 3:
to a mixture of 1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (70 mg,177.02umol,1 eq), 3- [ 6-fluoro-5- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (81.70 mg,177.02umol,1 eq) in MeOH (10 mL) and AcOH (2 mL) was added borane; 2-methylpyridine (37.87 mg,354.05umol,2 eq.) then the mixture was stirred at 25℃for 12 hours. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: xtime C18. Times.30 mm. 10um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 3- [ 6-fluoro-5- [4- [ [ 4-fluoro-1- [ [1- [ 6-fluoro-5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (41.4 mg,47.75umol,26.98% yield, 97% purity) as a white solid.
Exemplary Synthesis of Compound 194
Step 1:
at N 2 Pd (dppf) Cl2 (3.46 g,4.72mmol,0.05 eq.) and TEA (28.69 g,283.50mmol,39.46mL,3 eq.) were added to a solution of 2-bromo-4-methoxy-1-methyl-benzene (19 g,94.50mmol,1 eq.) in MeOH (50 mL). The suspension was degassed under vacuum and purged several times with CO. The mixture was stirred at 100℃under CO (3 MPa) for 96 hours. TLC showed the reaction was complete. The residue was filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (80 g,0% -3% (15 min) ethyl acetate/petroleum ether, 3% (15 min) ethyl acetate/petroleum ether) to give methyl 5-methoxy-2-methyl-benzoate (15.5 g,86.02mmol,91.02% yield) as a colorless oil.
Step 2:
to a solution of methyl 5-methoxy-2-methyl-benzoate (5 g,27.75mmol,1 eq.) in AcOH (50 mL) was added Br2 (8.87 g,55.49mmol,2.86mL,2 eq.). After the addition, the mixture was stirred at 20 ℃ for 2 hours. TLC showed the reaction was complete. The reaction mixture was taken up in saturated Na 2 S 2 O 3 (60 mL) was quenched and extracted with ethyl acetate (2X 40 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by chromatography on silica gel (80 g,0% -3% (20 min) ethyl acetate/petroleum ether, 3% (10 min) ethyl acetate Petroleum ether) to give methyl 4-bromo-5-methoxy-2-methyl-benzoate (1.1 g,3.64mmol,13.13% yield, 85.784% purity) as a white solid.
Step 3:
to a mixture of methyl 4-bromo-5-methoxy-2-methyl-benzoate (450 mg,1.74mmol,1 eq.) in DCE (10 mL) was added NBS (370.95 mg,2.08mmol,1.2 eq.) and AIBN (57.04 mg,347.36umol,0.2 eq.) at 20 ℃. The mixture was then stirred at 50℃for 16 hours. TLC showed a new main point. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (20 g,0% -3% (12 min) ethyl acetate/petroleum ether) to give methyl 4-bromo-2- (bromomethyl) -5-methoxy-benzoate (200 mg,591.73umol,34.07% yield) as a white solid.
Step 4:
to a mixture of methyl 4-bromo-2- (bromomethyl) -5-methoxy-benzoate (200 mg,591.73umol,1 eq.) and 3-aminopiperidine-2, 6-dione (113.73 mg,887.59umol,1.5 eq.) in MeCN (3 mL) was added DIEA (229.43 mg,1.78mmol,309.21ul,3 eq.). The mixture was stirred at 80℃for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by prep HPLC (column: xtime C18 x 40mm x 10um; mobile phase: [ water (0).225% FA)-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:20% -50%,10 min) to give 3- (5-bromo-6-methoxy-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (88 mg,249.17umol,42.11% yield) as a grey solid.
Step 5:
at N 2 Pd-PEPSI-pent Cl-o-picoline (21.43 mg,24.92umol,0.1 eq) and Cs2CO3 (162.37 mg,498.35umol,2 eq) were added to a solution of 3- (5-bromo-6-methoxy-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (88 mg,249.17umol,1 eq) and 4- (dimethoxymethyl) piperidine (79.35 mg,498.35umol,2 eq) in DMSO (5 mL). After addition, the mixture was stirred at 80℃under N 2 Stirred for 16 hours. LCMS showed the desired MS. The residue was filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -100% (20 min) ethyl acetate/petroleum ether, 0% -15% (10 min) methanol/dichloromethane, 15% (10 min) methanol/dichloromethane) to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl as a white solid]-6-methoxy-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (50 mg,63.73umol,25.58% yield, 55% purity).
Step 6:
to 3- [5- [4- (dimethoxymethyl) -1-piperidinyl]-6-methoxy-1-oxo-isoindolin-2-yl]To a solution of piperidine-2, 6-dione (50 mg,115.88umol,1 eq.) in THF (2 mL) was added HCl (2M, 972.11uL,16.78 eq.). After addition, the reaction was stirred at 20 ℃ for 16 hours. LCMS showed the desired MS. Addition of NaHCO to the residue 3 To adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 1- [2- (2, 6-dioxo-3-piperidinyl) -6-methoxy-1-oxo-isoindolin-5-yl as a white solid]Piperidine-4-carbaldehyde (55 mg, crude).
Step 7:
to 1- [2- (2, 6-dioxo-3-piperidyl) -6-methoxy-1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (55 mg,71.35umol,50% purity, 1 eq.) and 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Pyrimidin-4-yl]To a solution of 1H-indazole (35.13 mg,78.49umol,1.1 eq.) in MeOH (10 mL), THF (10 mL) and AcOH (1 mL) was added borane; 2-methylpyridine (15.26 mg,142.70umol,2 eq). After addition, the mixture was stirred at 20 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.1% TFA) -ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -50%,40 min) to give 3- [ 6-methoxy-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (22.3 mg,26.63umol,37.32% yield, 97.547% purity).
Exemplary Synthesis of Compound 195
Step 1:
to 6-fluoro-3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl]Piperazin-1-yl]Pyrimidin-4-yl]-5- (1-methylcyclopropoxy) -2H-indazole (85 mg,175.78umol,1 eq.) and 1- [2- (2, 6-dioxo- ]3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (65.63 mg,175.78umol,1 eq) was added borane in a mixture of THF (10 mL), meOH (10 mL) and AcOH (1 mL); 2-methylpyridine (37.60 mg,351.56umol,2 eq). The mixture was stirred at 20℃for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 6-fluoro-5- [4- [ [ 4-fluoro-4- [ [4- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (38.3 mg,45.47 mol,25.87% yield, 99.834% purity).
Exemplary Synthesis of Compound 196
Step 1:
to a solution of tert-butyl 4-fluoro-4- [ [ (2S) -2-methylpiperazin-1-yl ] methyl ] piperidine-1-carboxylate (109.78 mg,348.03umol,1.2 eq) and 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane (150 mg,348.03umol,1 eq) in DMSO (2 mL) was added TEA (105.65 mg,1.04mmol,145.32ul,3 eq). The reaction was then stirred at 90℃for 1 hour. LCMS showed that one peak with the desired mass was detected. The reaction mixture was diluted with water (15 mL) and extracted with dichloromethane (15 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 30% ethyl acetate/petroleum ether) to give 4-fluoro-4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylic acid tert-butyl ester (200 mg,262.83umol,75.52% yield, 93.3% purity) as a pale yellow oil.
Step 2:
to a mixture of tert-butyl 4-fluoro-4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylate (200 mg,281.70umol,1 eq.) in DCM (1 mL) and MeOH (1 mL) was added HCl/EtOAc (4 m,2mL,28.40 eq.). The mixture was stirred at 25℃for 1 hour. LCMS showed that one peak with the desired mass was detected. The mixture was concentrated at 45 ℃ under reduced pressure to give 4-fluoro-4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] piperidine-1-carboxylic acid tert-butyl ester (200 mg,262.83umol,75.52% yield, 93.3% purity) as a pale yellow oil.
Step 3:
3- [6- [ (3S) -4- [ (4-fluoro-4-piperidinyl) methyl]-3-methyl-piperazin-1-yl]Pyrimidin-4-yl]-5- (1-methylcyclopropoxy) -2H-indazole (38.90 mg,56.78 mol,70% purity, 1.07 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl)]A mixture of piperidine-4-carbaldehyde (25 mg,52.90umol,79% purity, 1 eq.) in HOAC (1 mL) and MeOH (10 mL) was stirred at 20deg.C for 20 min, followed by the addition of borane; 2-methylpyridine (11.32 mg, 105.80. Mu. Mol,2 eq). The mixture was then brought to 20℃under N 2 Stirred for 16 hours. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: xtime C18 x 30mM x 10um; mobile phase: [ water (10 mM NH 4 HCO 3 )-ACN];B%:10% -80%,40 min) to give 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-4- [ [ (2S) -2-methyl-4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl) as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (18.4 mg,21.61umol,40.85% yield, 98.3% purity).
Exemplary Synthesis of Compound 197
Compound 197 was prepared in a similar manner to compound 196 starting from 2- [ [3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl ] methoxy ] ethyl-trimethyl-silane.
Exemplary Synthesis of Compound 198
Step 1:
to a solution of 1- [2- (2, 6-dioxo-3-piperidinyl) -6-methoxy-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (90 mg,168.13 mol,72% purity, 1 eq) and 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] piperazin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (78.28 mg,168.13 mol,1 eq) in MeOH (10 mL), THF (10 mL) and AcOH (1 mL) was added borane; 2-methylpyridine (35.97 mg,336.26umol,2 eq). After addition, the mixture was stirred at 20 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18. Sup.100deg.C 30 mm. Sup.10um; mobile phase: [ water (0.1% TFA) -ACN ]; B%:0% -50%,40 min.) to afford 3- [5- [4- [ [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -6-methoxy-1-oxo-isoindol-2-yl ] piperidine-2, 6-dione as a white solid (31 mg,36.05umol,21.44% yield, 97.108% purity).
Exemplary Synthesis of Compound 199
Compound 199 was prepared in a similar manner to compound 190 using 1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde in the last step.
Exemplary Synthesis of Compound 200
Step 1:
to 4- (azetidin-3-yloxymethyl) -4-fluoro-piperidine-1-carboxylic acid tert-butyl ester (150 mg,520.19umol,1 eq.) and 2- [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl ]]Methoxy group]To a mixture of ethyl-trimethyl-silane (200 mg,464.03umol,8.92e-1 eq.) in DMSO (5 mL) was added TEA (52.64 mg,520.19umol,72.40uL,1 eq.). The mixture was stirred at 90℃for 16 hours. LCMS showed the desired MS. The residue was poured into water (30 mL). The aqueous phase was extracted with ethyl acetate (30 ml x 3). The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -40% (15 min) ethyl acetate/petroleum ether, 40% (10 min) ethyl acetate/petroleum ether) to give 4-fluoro-4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] as a brown solid ]Pyrimidin-4-yl]Azetidin-3-yl]Oxymethyl group]Tert-butyl piperidine-1-carboxylate (250 mg,343.05umol,65.95% yield, 93.707% purity).
Step 2:
to a mixture of tert-butyl 4-fluoro-4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] azetidin-3-yl ] oxymethyl ] piperidine-1-carboxylate (150 mg,219.65umol,1 eq.) in EtOAc (4 mL) was added HCl/EtOAc (4 m,4mL,72.84 eq.). The mixture was stirred at 20℃for 2 hours. TLC showed the reaction was complete. The residue was concentrated in vacuo to give 3- [6- [3- [ (4-fluoro-4-piperidinyl) methoxy ] azetidin-1-yl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (112 mg, crude, HCl) as a yellow solid.
Step 3:
to 3- [6- [3- [ (4-fluoro-4-piperidinyl) methoxy]Azetidin-1-yl]Pyrimidin-4-yl]-5- (1-methylcyclopropoxy) -2H-indazole (99 mg,218.77 mol,1.36 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (60 mg,160.70umol,1 eq) was added borane to a mixture of MeOH (10 mL), THF (10 mL) and AcOH (1 mL); 2-methylpyridine (34.38 mg, 321.39. Mu. Mol,2 eq.). The mixture was stirred at 40℃for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl) as a white solid]Pyrimidin-4-yl]Azetidin-3-yl]Oxymethyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (58 mg,70.71umol,44.00% yield, 98.738% purity).
Exemplary Synthesis of Compound 201
Step 1:
to 4-bromo-3-fluoro-2-methyl-benzoic acid methyl ester (2 g,8.10mmol,1 eq.) in CCl 4 AIBN (133 mg,0.81mmol,0.1 eq.) and NBS (1.44 g,8.10mmol,1 eq.) were added to a solution in (20 mL). The mixture was stirred at 80℃for 16 hours. LC-MS showed little starting material remaining and the desired compound was detected as the major product. The reaction mixture was partitioned between 30mL of water and 60mL (20 mL x 3) of DCM. The organic phase was separated over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=10/1 to 6/1). The compound 4-bromo-2- (bromomethyl) -3-fluoro-benzoic acid methyl ester (2.3 g,7.06mmol,87% yield) was obtained as a yellow solid.
Step 2:
to a solution of 3-aminopiperidine-2, 6-dione (284 mg,5.49mmol,7.78e-1 eq, HCl) in DMF (25 mL) was added DIEA (4.56 g,35.28mmol,6.15mL,5 eq) and stirred for 30 min followed by methyl 4-bromo-2- (bromomethyl) -3-fluoro-benzoate (2.3 g,7.06mmol,1 eq). The mixture was stirred at 100℃for 11.5 hours. LC-MS showed no residue of starting material and detected the desired compound as the major product. The reaction mixture was poured into cold water (60 mL) to form a precipitate. The mixture was filtered and the filter cake was dried in vacuo. The mixture was triturated with PE and methyl tert-butyl ether (V/v=1:1, 20 mL). The compound 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (1.4 g,4.10mmol,58% yield) was obtained as a grey solid.
/>
Step 3:
to 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dioneTo a solution of (300 mg,0.88mmol,1 eq), 4- (dimethoxymethyl) piperidine (168 mg,1.06mmol,1.2 eq.) in DMSO (6 mL) was added Cs2CO3 (573 mg,1.76mmol,2 eq.) and PD-PEPSI (86 mg,0.088mmol,0.1 eq.). The mixture was stirred at 100℃for 12h. LC-MS showed detection of the desired compound. The reaction mixture was partitioned between 6mL of water and 36mL of EtOAc (12 mL x 3). The organic phase was separated over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography by preparative TLC (SiO 2, DCM: meoh=10:1). The compound 3- [5- [4- (dimethoxymethyl) -1-piperidinyl was obtained as a yellow solid]-4-fluoro-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (130 mg,0.31mmol,35% yield).
Step 4:
to a solution of 3- [5- [4- (dimethoxymethyl) -1-piperidinyl ] -4-fluoro-1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (80 mg,0.19mmol,1 eq.) in THF (0.5 mL) was added HCl/dioxane (4 m,2mL,41.94 eq.). The mixture was stirred at 25℃for 1 hour. TLC plates showed starting material was consumed. The reaction mixture was concentrated in vacuo to give a residue. The compound 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (70 mg,0.19mmol,98% yield) was obtained as a yellow solid.
Step 5:
to a solution of 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -1H-pyrazolo [3,4-c ] pyridine (43 mg,0.089mmol,1 eq, HCl) in MeOH (2 mL) was added NaOAc (36 mg,0.44mmol,5 eq), the mixture was stirred at 25 ℃ for 10min, followed by 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (33 mg,0.089mmol,1 eq), acOH (0.05 mL) and stirring was continued for 20min. 2-methyl-pyridine borane (28 mg,0.27mmol,3 eq.) was then added. The mixture was stirred at 25℃for 15.5 hours. LC-MS showed no residue of starting material. The desired compound was detected as the major product. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: phenomenex Synergi C18 150 x 25mm x 10um; mobile phase: [ water (0.225% FA) -ACN ]; B%:3% -33%,10 min). The compound 3- (4-fluoro-5- (4- ((4- ((1- (6- (5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] pyridin-3-yl) pyrimidin-4-yl) piperidin-4-yl) methyl) piperazin-1-yl) methyl) piperidin-1-oxoisoindol-2-yl) piperidine-2, 6-dione (35.1 mg,0.044mmol,49% yield) was obtained as a white solid.
Exemplary Synthesis of Compound 202
Step 1:
to 4- [ (4-formylcyclohexyl) methyl]To a mixture of tert-butyl piperazine-1-carboxylate (170 mg,547.63umol,1 eq) in MeOH (3 mL) and HOAc (0.5 mL) was added a solution of 3- (4-fluoro-1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (370 mg,803.69umol,1.47 eq, TFA) and DIEA (70.78 mg,547.63umol,95.38ul,1 eq) in MeOH (1 mL). Then borane is added; 2-methylpyridine (117.15 mg,1.10mmol,2 eq.) was added to the above solution. After the addition, the reaction solution was stirred at 20 ℃ for 12h. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove most of the solvent. The residue was purified by prep HPLC (column: xtime C18 x 40mm x 10um; mobile phase: [ water (0.05% NH) 3 H 2 O)-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:45% -75%,10 min) to give 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a white solid]Piperazin-1-yl]Methyl group]Cyclohexyl group]Methyl group]Piperazine-1-carboxylic acid tert-butyl ester (137 mg, 183).87umol,33.58% yield, 86% purity).
Step 2:
to a solution of tert-butyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] cyclohexyl ] methyl ] piperazine-1-carboxylate (157 mg,245.01umol,1 eq.) in DCM (3 mL) was added TFA (27.94 mg,245.01umol,18.14uL,1 eq.). After the addition, the reaction solution was stirred at 20 ℃ for 1 hour. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 3- [ 4-fluoro-1-oxo-5- [4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ] methyl ] piperazin-1-yl ] isoindolin-2-yl ] piperidine-2, 6-dione (180 mg, crude, TFA) as a brown solid. The crude product was used directly in the next step.
Step 3:
to a solution of 3- [ 4-fluoro-1-oxo-5- [4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ] methyl ] piperazin-1-yl ] isoindolin-2-yl ] piperidine-2, 6-dione (90 mg,137.47 mol,1.15 eq, TFA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (36 mg,119.70 mol,1 eq) in DMSO (2 mL) was added TEA (121.13 mg,1.20mmol,166.61ul,10 eq). After the addition, the reaction solution was stirred at 90 ℃ for 4 hours. LCMS showed the reaction was complete. The reaction mixture was diluted with water (10 mL) and extracted with dichloromethane (10 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: xtime C18. Times.30 mm. Times.10 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,40 min) to afford 3- [ 4-fluoro-5- [4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] cyclohexyl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (29.9 mg,36.71umol,30.67% yield, 98.83% purity) as a pale yellow solid.
Exemplary Synthesis of Compound 203
Compound 203 was prepared in a similar manner to compound 202 using 3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) -1H-indazole in the last step.
Exemplary Synthesis of Compound 204
Step 1:
to a solution of methyl 4-formylcyclohexane carboxylate (3 g,17.63mmol,1 eq.) and tert-butyl piperazine-1-carboxylate (3.28 g,17.63mmol,1 eq.) in MeOH (30 mL) and HOAc (3 mL) was added borane; 2-methylpyridine (3.77 g,35.25mmol,2 eq.). After the addition, the reaction solution was stirred at 20 ℃ for 12 hours. TLC (petroleum ether: ethyl acetate=1:1) showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove most of the solvent. The residue was purified by silica gel column chromatography (0% to 60% ethyl acetate/petroleum ether) to give tert-butyl 4- [ (4-methoxycarbonylcyclohexyl) methyl ] piperazine-1-carboxylate (4 g,11.75mmol,66.66% yield) as a colorless oil.
/>
Step 2:
at 0 ℃ at N 2 Downward 4- [ (4-methoxycarbonylcyclohexyl) methyl group]To a solution of tert-butyl piperazine-1-carboxylate (4 g,11.75mmol,1 eq.) in THF (50 mL) was added LiAlH4 (535.05 mg,14.10mmol,1.2 eq.) in portions. After the addition, the reaction mixture was stirred at 0 ℃ for 1 hour. TLC (petroleum ether: ethyl acetate=0:1) showed the reaction was complete. By H 2 O (1 mL), followed by 15% aqueous NaOH (1 mL) and H 2 O (3 mL) to quench the reaction mixture And (3) an object. After stirring at room temperature for 0.5 hours, the solids were removed by filtration. The filtrate was concentrated to dryness to give the crude product. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give 4- [ [4- (hydroxymethyl) cyclohexyl ] as a colorless oil]Methyl group]Piperazine-1-carboxylic acid tert-butyl ester (2.3 g,7.36mmol,62.65% yield, 100% purity).
Step 3:
at N 2 Atmospheric direction to 4- [ [4- (hydroxymethyl) cyclohexyl)]Methyl group]To a solution of tert-butyl piperazine-1-carboxylate (220 mg,704.12umol,1 eq.) in DCM (5 mL) was added DMP (597.29 mg,1.41mmol,435.98uL,2 eq.). The mixture was stirred at 30℃for 2 hours. TLC showed complete consumption of starting material. Pouring the residue into NaHCO 3 To adjust ph=7-8, and Na is added 2 SO 3 (20 mL). The aqueous phase was extracted with DCM (30 ml x 3). The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 4- [ (4-formylcyclohexyl) methyl as a colourless oil]Tert-butyl piperazine-1-carboxylate (202 mg,649.71umol,92.27% yield, 99.846% purity).
Step 4:
to 4- [ (4-formylcyclohexyl) methyl]Tert-butyl piperazine-1-carboxylate (200 mg,644.27umol,1 eq) and 3- (6-fluoro-1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (223.15 mg,644.27umol,1 eq) in a mixture of MeOH (10 mL) and AcOH (1 mL) was added borane; 2-methylpyridine (137.82 mg,1.29mmol,2 eq.). The mixture was stirred at 40℃for 16 hours. LCMS showed the desired MS. The residue was poured into water (30 mL). The aqueous phase was extracted with ethyl acetate (30 ml x 3). Will be combined The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -20%,40 min) to give 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] as a white solid]Piperazin-1-yl]Methyl group]Cyclohexyl group]Methyl group]Tert-butyl piperazine-1-carboxylate (190 mg,291.96umol,45.32% yield, 98.465% purity).
Step 5:
to a mixture of tert-butyl 4- [ [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] cyclohexyl ] methyl ] piperazine-1-carboxylate (180 mg,280.90umol,1 eq.) in DCM (2 mL) was added TFA (32.03 mg,280.90umol,20.80ul,1 eq.). The mixture was stirred at 35℃for 1 hour. LCMS showed the desired MS. The residue was concentrated in vacuo to give 3- [ 6-fluoro-1-oxo-5- [4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ] methyl ] piperazin-1-yl ] isoindolin-2-yl ] piperidine-2, 6-dione (190 mg, crude, TFA) as a brown gum.
Step 6:
to 3- [ 6-fluoro-1-oxo-5- [4- [ [4- (piperazin-1-ylmethyl) cyclohexyl ]Methyl group]Piperazin-1-yl]Isoindolin-2-yl]To a solution of piperidine-2, 6-dione (75 mg,138.72umol,1 eq) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1H-indazole (41.72 mg,138.72umol,1 eq) in DMSO (2 mL) was added TEA (14.04 mg,138.72umol,19.31uL,1 eq). After addition, the mixture was stirred at 90 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was purified with ethyl acetate (20 mL)* 3) And (5) extracting. The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 6-fluoro-5- [4- [ [4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]Cyclohexyl group]Methyl group]Piperazin-1-yl]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (31.8 mg,39.40umol,28.40% yield, 99.734% purity).
Exemplary Synthesis of Compound 205
Compound 205 was prepared in a similar manner to compound 204 using 3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) -1H-indazole in the last step.
Exemplary Synthesis of Compound 206
Step 1:
to a mixture of 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (70 mg,150.67umol,1 eq), 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (56.26 mg,150.67umol,1 eq.) in MeOH (10 mL) and AcOH (2 mL) was added borane; 2-methylpyridine (48.35 mg, 452.01. Mu. Mol,3 eq.) then the mixture was stirred at 25℃for 12 hours. LCMS showed the desired MS. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: xtime C18. Times.30 mm. Times.10 um; mobile phase: [ water (10 mM NH4HCO 3) -ACN ]; B%:10% -80%,40 min) to afford 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (43.4 mg,52.27 mol,34.69% yield, 99% purity) as a white solid.
Exemplary Synthesis of Compound 207
Step 1:
to 4-fluoro-4- (4-piperidinylmethyl) piperidine-1-carboxylic acid tert-butyl ester (66.91 mg,222.72umol,1 eq.) 2- [ [3- (6-chloropyrimidin-4-yl) -6-fluoro-5- (1-methylcyclopropoxy) indazol-2-yl ]Methoxy group]To a mixture of ethyl-trimethyl-silane (100 mg,222.72umol,1 eq.) in DMSO (5 mL) was added DIEA (287.85 mg,2.23mmol,387.94uL,10 eq.) and the mixture was stirred at 100deg.C under N2 atmosphere for 12 hours. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=3:1) showed a new major spot. Pouring the obtained product into H 2 O (10 mL). The mixture was extracted with ethyl acetate (50 ml x 3). The organic phase was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by silica gel column chromatography (0% to 27% ethyl acetate/petroleum ether) to give 4-fluoro-4- [ [1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] as a yellow oil]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Tert-butyl piperidine-1-carboxylate (100 mg,129.04umol,57.94% yield, 92% purity).
Step 2:
to a mixture of tert-butyl 4-fluoro-4- [ [1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperidine-1-carboxylate (100 mg,140.26umol,1 eq) in MeOH (10 mL) was added HCl/EtOAc (4 m,4mL,114.07 eq), and the mixture was stirred at 25 ℃ under an atmosphere of N2 for 2 hours. TLC (petroleum ether: ethyl acetate=3:1) showed a new major spot. The resulting product was concentrated in vacuo to give 6-fluoro-3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (70 mg, crude, HCl) as a yellow oil.
Step 3:
to a mixture of 6-fluoro-3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ] -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -2H-indazole (40 mg,82.89umol,1 eq), 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (30.95 mg,82.89umol,1 eq.) in MeOH (10 mL) and AcOH (2 mL) was added borane; 2-methylpyridine (26.60 mg,248.67umol,3 eq.) then the mixture was stirred at 25℃for 12 hours. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: xtime C18. Times.30 mm. 10um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -35%,40 min) to afford 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-4- [ [1- [6- [ 6-fluoro-5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (31.5 mg,36.75umol,44.34% yield, 98% purity) as a white solid.
Exemplary Synthesis of Compound 208
Step 1:
to 3- (6-fluoro-1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (300 mg,866.16umol,1 eq.) and 4- [4- (p-toluenesulfonyloxymethyl) cyclohexyloxy ]Piperidine-1-carboxylic acid tert-butyl ester (486.04 mg,1.04mmol,1.2 eq.) in CH 3 KI (1.44 g,8.66mmol,10 eq.) and DIEA (1.12 g, 8.66)mmol,1.51ml,10 eq) followed by stirring the mixture at 90 ℃ for 16 hours. LCMS showed 77.4% of the desired compound and TLC (petroleum ether: ethyl acetate=0:1, uv=254 nm) showed the formation of new spots. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel20gSilica gel flash column, eluent: 0% -95% ethyl acetate/petroleum ether gradient, 50 mL/min). Obtaining the compound 4- [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] as a pale yellow solid]Piperazin-1-yl]Methyl group]Cyclohexyloxy radical]Piperidine-1-carboxylic acid tert-butyl ester (0.7 g, crude).
Step 2:
to a solution of tert-butyl 4- [4- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] cyclohexyloxy ] piperidine-1-carboxylate (350 mg,545.37 mol,1 eq) in DCM (3 mL) was added TFA (1.54 g,13.51mmol,1mL,24.77 eq), and the mixture was stirred at 25℃for 2 h. TLC (dichloromethane: methanol=10:1, uv=254 nm) showed the formation of new spots. The reaction mixture was concentrated under reduced pressure to give a residue. The compound 3- [ 6-fluoro-1-oxo-5- [4- [ [4- (4-piperidinyloxy) cyclohexyl ] methyl ] piperazin-1-yl ] isoindolin-2-yl ] piperidine-2, 6-dione (356 mg,376.23umol,68.99% yield, 69.1% purity, TFA) was obtained as a pale yellow oil.
Step 3:
to a solution of 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (60 mg,199.51umol,1 eq) and 3- [ 6-fluoro-1-oxo-5- [4- [ [4- (4-piperidinyloxy) cyclohexyl ] methyl ] piperazin-1-yl ] isoindolin-2-yl ] piperidine-2, 6-dione (170.06 mg,259.36umol,1.3 eq, TFA) in DMSO (5 mL) was added DIEA (128.92 mg,997.53umol,173.75uL,5 eq) followed by stirring the solution at 90℃for 16 hours. LCMS showed about 30% of the desired compound and the starting material was completely consumed. The residue was purified by preparative HPLC (column: xtimate C18 x 30mm x 10um; mobile phase: [ water (0.225% fa) -ACN ]; B%:0% -40%,40 min). The compound 3- [ 6-fluoro-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclohexyl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (17.6 mg,20.80umol,10.43% yield, 95.262% purity) was obtained as a white solid.
Exemplary Synthesis of Compound 209
Step 1:
at N 2 Downward 4- [ (4-fluoro-4-piperidinyl) methyl]To a solution of benzyl piperazine-1-carboxylate (350 mg,941.17 mol,1.28 eq, HCl) and 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (250 mg,732.85 mol,1 eq) in DMSO (5 mL) was added Pd-PEPPSI-pentcl-o-methylpyridine (39.79 mg,73.29 mol,0.1 eq) and Cs2CO3 (477.56 mg,1.47mmol,2 eq). After addition, the mixture was stirred at 80 ℃ under N2 for 16 hours. LCMS showed the desired MS. The residue was filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (12 g,0% -100% (20 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a white solid ]-4-fluoro-4-piperidinyl]Methyl group]Benzyl piperazine-1-carboxylate (130 mg,96.56umol,13.18% yield, 44.242% purity).
/>
Step 2:
to a mixture of 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-fluoro-4-piperidinyl ] methyl ] piperazine-1-carboxylic acid benzyl ester (130 mg,218.25umol,1 eq.) was added TFA (3.08 g,27.01mmol,2ml,123.76 eq.). The mixture was stirred at 70℃for 1 hour. TLC showed the reaction was complete. The residue was concentrated in vacuo to give 3- [ 4-fluoro-5- [ 4-fluoro-4- (piperazin-1-ylmethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (130 mg, crude, TFA) as a brown gum.
Step 3:
to 3- [ 4-fluoro-5- [ 4-fluoro-4- (piperazin-1-ylmethyl) -1-piperidinyl]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (100 mg,216.68umol,1 eq.) and 1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl]Pyrimidin-4-yl]To a solution of piperidine-4-carbaldehyde (81.78 mg,216.68umol,1 eq.) in MeOH (10 mL) and AcOH (1 mL) was added borane; 2-methylpyridine (46.35 mg, 433.37. Mu. Mol,2 eq.). After addition, the mixture was stirred at 20 ℃ for 16 hours. LCMS showed the desired MS. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: 3_Phenomenex Luna C18 75*30mm*3um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 4-fluoro-5- [ 4-fluoro-4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (40 mg,47.65 u)mol,21.99% yield, 98.028% purity).
Exemplary Synthesis of Compound 210
Step 1:
to a solution of cyclohexane-1, 4-diol (4.2 g,36.16mmol,1.00 eq.) in tetrahydrofuran (80 mL) was added triethylamine (8.42 g,83.16mmol,2.30 eq.) and chloro (trimethyl) silane (8.64 g,79.55mmol,2.20 eq.) at 0 ℃ and the reaction mixture was stirred at 25 ℃ for 0.5 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. Triethylsilane (14.72 g,126.55mmol,3.50 eq.) and trimethyltrisilane triflate (12.05 g,54.24mmol,1.50 eq.) were then added dropwise to a stirred solution of the above residue and benzyl 4-oxopiperidine-1-carboxylate (25.30 g,108.47mmol,3.00 eq.) in dichloromethane (80 mL) at-60 ℃ and the reaction mixture stirred under nitrogen for 12 hours at 25 ℃. The reaction mixture was diluted with 100mL of water and extracted with dichloromethane (100 mL. Times.2). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by semi-preparative reverse phase HPLC (70% -90% acetonitrile +0.225% formic acid/water, 20 min). The compound 4- [4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] cyclohexyloxy ] piperidine-1-carboxylic acid benzyl ester (5.3 g,9.62mmol,26% yield) was obtained as a white solid.
Step 2:
to a solution of benzyl 4- [4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] cyclohexyloxy ] piperidine-1-carboxylate (5.3 g,9.62mmol,1.00 eq.) in methanol (100 mL) under nitrogen was added a palladium on activated carbon catalyst (1.0 g,9.62mmol,10% purity, 1.00 eq.) and ammonium hydroxide (0.1 mL,25% purity). The suspension was degassed and purged 3 times with hydrogen. The mixture was stirred at 25℃under hydrogen (15 Psi) for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give 4- [4- (4-piperidinyloxy) cyclohexyloxy ] piperidine (2.6 g,9.21mmol,95% yield) as a white solid.
Step 3:
to a solution of 4- [4- (4-piperidinyloxy) cyclohexyloxy ] piperidine (700 mg,2.48mmol,1.97 eq) and 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (430 mg,1.26mmol,1.00 eq) in dimethyl sulfoxide (7 mL) under nitrogen was added cesium carbonate (823mg, 2.52mmol,2.00 eq) and 1, 3-bis [2, 6-bis (1-ethylpropyl) phenyl ] -2H-imidazole; 3-chloropyridine; palladium dichloride (100 mg,0.12mmol,0.10 eq.). The mixture was stirred at 100 ℃ under nitrogen for 12 hours. The reaction mixture was filtered to give a black liquid. The liquid was then purified by semi-preparative reverse phase HPLC (9% -39% acetonitrile +0.1% trifluoroacetic acid/water for 11 min). The compound 3- [ 4-fluoro-1-oxo-5- [4- [4- (4-piperidinyloxy) cyclohexoxy ] -1-piperidinyl ] isoindolin-2-yl ] piperidine-2, 6-dione (80 mg,0.12mmol,9% yield, trifluoroacetate) was obtained as a white solid.
/>
Step 4:
to a solution of 3- [ 4-fluoro-1-oxo-5- [4- [4- (4-piperidinyloxy) cyclohexyloxy ] -1-piperidinyl ] isoindolin-2-yl ] piperidine-2, 6-dione (40 mg,0.06mmol,1.00 eq., trifluoroacetate) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine (33 mg,0.06mmol,1.00 eq.) in dimethyl sulfoxide (1 mL) was added N, N-diisopropylethylamine (23 mg,0.18mmol,3.00 eq.). The mixture was stirred at 100℃for 0.5 h. The reaction mixture was diluted with 20mL of dichloromethane and washed with saturated ammonium chloride solution, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 3- [ 4-fluoro-5- [4- [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclohexoxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (63 mg,0.06mmol,98% yield) as a white solid.
Step 5:
to a solution of 3- [ 4-fluoro-5- [4- [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclohexoxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (63 mg,0.06mmol,1.00 eq.) in hydrogen chloride/dioxane (4 m,1.5mL,100 eq.) was added water (0.3 mL). The mixture was stirred at 25℃for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by semi-preparative reverse phase HPLC (28% -58% acetonitrile +0.225% formic acid/water, 7 min). The compound 3- [ 4-fluoro-5- [4- [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclohexoxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (18.0 mg,0.02mmol,34% yield, 97% purity, formate) was obtained as an off-white solid.
Exemplary Synthesis of Compound 211
Step 1:
to a solution of 2-methylpropan-2-amine (8.80 g,120.36mmol,12.65mL,1 eq.) in DCM (50 mL) at-70℃was added dropwise Br 2 (19.23 g,120.36mmol,6.20mL,1 eq.) in DCM (50 mL), anThe mixture was stirred at-70℃for 1 hour. A solution of 3-hydroxy-2-methyl-benzoic acid methyl ester (20 g,120.36mmol,1 eq.) in DCM (200 mL) was then added dropwise and the resulting mixture was warmed to 20℃and stirred for 8 hours. TLC (petroleum ether: ethyl acetate=5:1) indicated complete consumption of the reactants. The mixture was quenched with water (100 mL), extracted with DCM (500 mL), washed with brine (500 mL), and dried over Na 2 SO 4 Dried and concentrated under reduced pressure. Purification by silica gel chromatography (0% -3% EtOAc/petroleum ether) afforded 4-bromo-3-hydroxy-2-methyl-benzoic acid methyl ester (7.77 g,31.71mmol,26.34% yield) as a yellow solid.
Step 2:
to a solution of methyl 4-bromo-3-hydroxy-2-methyl-benzoate (7.77 g,31.71mmol,1 eq.) in MeCN (70 mL) was added MeI (13.50 g,95.12mmol,5.92mL,3 eq.) and K2CO3 (5.26 g,38.05mmol,1.2 eq.). The mixture was stirred at 50℃for 5 hours. TLC (petroleum ether: ethyl acetate=20:1) indicated complete consumption of the reactants. The mixture was quenched with water (10 mL), extracted with EtOAc (50 mL), washed with brine (50 mL) and dried over Na 2 SO 4 Dried and concentrated under reduced pressure. Purification by silica gel chromatography (0% -3% etoac/petroleum ether) afforded 4-bromo-3-methoxy-2-methyl-benzoic acid methyl ester (7.44 g,28.72mmol,90.57% yield) as a yellow oil.
Step 3:
to a stirred solution of methyl 4-bromo-3-methoxy-2-methyl-benzoate (2 g,7.72mmol,1 eq.) in DCE (10 mL) under nitrogen atmosphere was added NBS (1.65 g,9.26mmol,1.2 eq.) followed by 2- [ (E) - (1-cyano-1-methyl-ethyl) azo ] -2-methyl-propionitrile (63.38 mg,385.96umol,0.05 eq.) and the resulting mixture was vigorously stirred at 80 ℃ for 1 hour to give a yellow solution. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel chromatography (10% ethyl acetate/petroleum ether) to give methyl 4-bromo-2- (bromomethyl) -3-methoxy-benzoate (2.3 g,6.80mmol,88.16% yield) as a yellow oil.
Step 4:
to a mixture of methyl 4-bromo-2- (bromomethyl) -3-methoxy-benzoate (2.3 g,6.80mmol,1 eq.) and 3-aminopiperidine-2, 6-dione (1.34 g,8.17mmol,1.2 eq., HCl) in DMF (10 mL) at 20 ℃ was added DIEA (4.40 g,34.02mmol,5.93mL,5 eq.) in one portion. The mixture was stirred at 85 ℃ for 48 hours. LCMS showed the desired MS. The crude product was concentrated in vacuo. The crude product was purified by MeCN (20 mL) and H 2 O (20 mL) was ground at 20deg.C. The crude was filtered and the solid concentrated in vacuo to give 3- (5-bromo-4-methoxy-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (2 g,5.66mmol,83.22% yield) as a dark grey solid.
Step 5:
to a solution of 3- (5-bromo-4-methoxy-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (500 mg,1.42mmol,1 eq.) and 4- (dimethoxymethyl) piperidine (450.85 mg,2.83mmol,2 eq.) in DMF (5 mL) was added Cs 2 CO 3 (922.57 mg,2.83mmol,2 eq.) and Pd-PEPSI-pent Cl-o-methylpyridine (121.76 mg,141.58umol,0.1 eq.). After addition, the reaction mixture was taken up in N at 80 ℃ 2 Stirred for 12 hours. LCMS (EB 16-1785-P1A 1) showed the desired MS. The mixture was cooled to 20 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). Will be combinedThe organic phase was washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -100% (10 min) ethyl acetate/petroleum ether, 100% (10 min) ethyl acetate/petroleum ether) to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl as a yellow solid]-4-methoxy-1-oxo-isoindolin-2-yl ]Piperidine-2, 6-dione (120 mg,183.55umol,12.97% yield, 66% purity).
Step 6:
at 20 ℃ at N 2 Downward 3- [5- [4- (dimethoxymethyl) -1-piperidinyl]-4-methoxy-1-oxo-isoindolin-2-yl]To a mixture of piperidine-2, 6-dione (170.00 mg,393.99umol,1 eq.) in THF (5 mL) was added HCl (2M, 3.17mL,16.10 eq.) in one portion. The mixture was stirred at 20 ℃ for 1 hour to give a yellow solution. LCMS showed the desired MS. Pouring the residue into saturated NaHCO 3 To adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 1- [2- (2, 6-dioxo-3-piperidinyl) -4-methoxy-1-oxo-isoindolin-5-yl as a yellow solid]Piperidine-4-carbaldehyde (140 mg,247.01umol,62.69% yield, 68% purity).
Step 7:
at 20 ℃ at N 2 Downward 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Pyrimidin-4-yl]-2H-indazole (55.28 mg,123.50umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -4-methoxy-1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (70.00 mg,123.50umol,68% purity, 1 eq.) in MeOH (10 mL) Adding borane at one time; 2-methylpyridine (26.42 mg, 247.01. Mu.mol, 2 eq.) and HOAc (7.42 mg, 123.50. Mu.mol, 7.06. Mu.L, 1 eq.). The mixture was stirred at 20 ℃ for 2 hours to give a yellow solution. LCMS showed the desired MS. Pouring the obtained product into H 2 O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -40%,40 min) to give 3- [ 4-methoxy-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (58.8 mg,71.69umol,58.05% yield, 99.61% purity, FA).
Exemplary Synthesis of Compound 212
Step 1:
at 20 ℃ at N 2 Downward 3- [6- [4- [ (4-fluoro-4-piperidinyl) methyl ]]Piperazin-1-yl]Pyrimidin-4-yl]-5- (1-methylcyclopropoxy) -2H-indazole (57.50 mg,123.50umol,1 eq.) and 1- [2- (2, 6-dioxo-3-piperidinyl) -4-methoxy-1-oxo-isoindolin-5-yl ]To a mixture of piperidine-4-carbaldehyde (70.00 mg,123.50umol,68% purity, 1 eq.) in MeOH (10 mL) was added borane in one portion; 2-methylpyridine (26.42 mg, 247.01. Mu.mol, 2 eq.) and HOAc (7.42 mg, 123.50. Mu.mol, 7.06. Mu.L, 1 eq.). The mixture was stirred at 20 ℃ for 10 hours to give a yellow solution. LCMS showed the desired MS. Pouring the obtained product into H 2 O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)];B%:0% -35%,40 min) to give 3- [5- [4- [ [ 4-fluoro-4- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]Piperazin-1-yl]Methyl group]-1-piperidinyl group]Methyl group]-1-piperidinyl group]-4-methoxy-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (46.7 mg,53.25umol,43.12% yield, 95.21% purity, FA).
Exemplary Synthesis of Compound 213
Step 1:
to 3- [ 4-fluoro-1-oxo-5- [4- [ [4- (4-piperidinyloxy) cyclohexyl ]]Methyl group]Piperazin-1-yl]Isoindolin-2-yl]Piperidine-2, 6-dione (252.15 mg,465.51umol,1 eq.) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (140 mg,465.51umol,1 eq.) were added to a mixture of piperidine-2, 6-dione in DMSO (5 mL), followed by stirring the mixture at 80℃under an atmosphere of N2 for 12 hours. LCMS showed the desired MS. Pouring the obtained product into H 2 O (10 mL). The mixture was extracted with ethyl acetate (50 ml x 3). The organic phase was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by preparative HPLC (column: xtime C18. Times.40 mm. Times.10 um; mobile phase: [ water (FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:10% -40%,10 min) to give 3- [ 4-fluoro-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] as a white solid]Pyrimidin-4-yl]-4-piperidinyl]Oxy group]Cyclohexyl group]Methyl group]Piperazin-1-yl]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (88.6 mg,107.74umol,23.14% yield, 98% purity).
Exemplary Synthesis of Compound 214
Step 1:
to a solution of 3, 6-difluorophthalic acid (1.5 g,7.42mmol,1 eq.) and 3-aminopiperidine-2, 6-dione (1.83 g,11.13mmol,1.5 eq., HCl) in HOAc (20 mL) was added NaOAc (1.83 g,22.26mmol,3 eq.) followed by stirring at 120℃for 1h. LCMS showed the reaction was complete. The reaction mixture was poured into water (50 mL), followed by filtration, and the filter cake was washed with water (20 ml×3), followed by concentration to give a crude product, which was then triturated with methyl tert-butyl ether (26 mL), followed by concentration to give a residue. The residue was used in the next step without further purification. The product 2- (2, 6-dioxo-3-piperidyl) -4, 7-difluoro-isoindoline-1, 3-dione (1.88 g, crude product) was a brown solid.
Step 2:
to a solution of 2- (2, 6-dioxo-3-piperidinyl) -4, 7-difluoro-isoindoline-1, 3-dione (2.18 g,7.41mmol,1 eq.) and tert-butyl 4- (4-piperidinylmethyl) piperazine-1-carboxylate (2.10 g,7.41mmol,1 eq.) in DMSO (10 mL) was added DIEA (2.87 g,22.23mmol,3.87mL,3 eq.) followed by stirring at 95℃for 1h. LCMS showed the reaction was complete. The reaction mixture was acidified to pH 5-6 by formic acid, then poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 2). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=0/1) to give 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -7-fluoro-1, 3-dioxo-isoindolin-4-yl ] as a yellow solid]-4-piperidinyl]Methyl group]Piperazine-1-carboxylic acid tert-butyl ester (2.6 g,4.57mmol,61.67% yield, 98% purity).
Step 3:
to a solution of tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -7-fluoro-1, 3-dioxo-isoindolin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (2.5 g,4.48mmol,1 eq.) in HOAc (36 mL) was added Zn (11.5 g,175.87mmol,39.23 eq.). The mixture was stirred at 90℃for 3h. LCMS showed the reaction was complete. The reaction mixture was filtered, and the filtrate was concentrated to give a residue, followed by addition of water (60 mL), followed by addition of saturated aqueous sodium bicarbonate (100 mL), and the mixture was extracted with ethyl acetate (80 ml×3). The combined organic layers were washed with brine (40 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a residue. The residue was purified by preparative HPLC (YMC Triart C18.50 mm.7 um; mobile phase: [ water (0.225% FA) -ACN ]; B%:5% -35%,20 min) to give tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -7-fluoro-1-oxo-isoindol-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (440 mg,809.38umol,18.05% yield) as a gray solid.
Step 4:
at 25 ℃ at N 2 Down 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -7-fluoro-1-oxo-isoindolin-4-yl ]]-4-piperidinyl]Methyl group]To a mixture of tert-butyl piperazine-1-carboxylate (80 mg,147.16 mol,1 eq.) in DCM (2 mL) was added TFA (1.23 g,10.80mmol,800.00uL,73.42 eq.) in one portion. The mixture was stirred at 25℃for 1 hour. LCMS showed the reaction was complete. The mixture was concentrated at 40 ℃ under reduced pressure. The residue was used in the next step without further purification. Product 3- [ 7-fluoro-1-oxo-4- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Isoindolin-2-yl]Piperidine-2, 6-dione (60 mg,129.87umol,88.25% yield, 96% purity) was as a grey solid.
Step 5:
at 25 ℃ at N 2 Downward 3- [ 7-fluoro-1-oxo-4- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Isoindolin-2-yl]Piperidine-2, 6-dione (60 mg,135.28umol,1 eq.) and 1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl]Pyrimidin-4-yl]Piperidine-4-carbaldehyde (51.06 mg,135.28umol,1 eq.) was added in one portion to a mixture of MeOH (4 mL) and HOAc (0.4 mL); 2-methylpyridine (28.94 mg,270.57 mol,2 eq). The mixture was stirred at 25℃for 16 hours. LCMS showed the reaction was complete. The mixture was concentrated at 40 ℃ under reduced pressure. The residue was purified by prep HPLC (Xtimate C18100 x 30mm x 10um; mobile phase: [ water (0.225% FA) -ACN ]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -30%,40 min) to give 3- [ 7-fluoro-4- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] as a gray solid]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (24.3 mg,28.47 mol,21.04% yield, 99.7% purity, FA).
Exemplary Synthesis of Compound 215
Step 1:
a solution of 2-methoxy-6-methyl-aniline (25 g,182.24mmol,1 eq.) in MeOH (100 mL) and AcOH (30 mL). The flask was cooled to 0deg.C and Br in AcOH (30 mL) 2 (29.12 g,182.24mmol,9.39mL,1 eq.) was added drop wise to the reaction. The reaction mixture was stirred at 0 ℃ for 2 hours, then warmed to 20 ℃ for 1 hour. TLC (petroleum ether: ethyl acetate=10:1) showed a new spot. The reaction mixture was concentrated. 15% NaOH was added at 0deg.C to neutralize the reactants. Ethyl acetate (100 mL) was added to extract the reaction mixture. The organic layer was concentrated after drying over anhydrous sodium sulfate to give 4-bromo-2-methoxy-6-methyl-aniline (35 g,161.98mmol,88.88% yield) as a brown oil.
Step 2:
the synthesized 4-bromo-2-methoxy-6-methyl-aniline (30 g,138.84mmol,1 eq) was suspended in concentrated aqueous HCl (12 m,85.71ml,7.41 eq) and ice water (32.52 g,1.81mol,13 eq) cooled to 0 ℃. Drop wise NaNO addition 2 (10.06 g,145.79mmol,1.05 eq.) in H2O (200 mL). The resulting mixture was stirred at 0deg.C for 30min and taken up with Na 2 CO 3 The aqueous solution (14.72 g,138.84mmol,1 eq.) was neutralized. The initial diazonium salt mixture was then added to CuCN (14.92 g,166.62mmol,36.40mL,1.20 eq.) at H 2 O (100 mL) and NaCN (20.40 g,416.27mmol,3.00 eq.) in H 2 To the suspension in O (100 mL), toluene (200 mL) was then added, and the mixture was stirred at 0deg.C for 1h, at 25deg.C for 2h, and at 50deg.C for 1h. TLC (petroleum ether: ethyl acetate=10:1, rf=0.63) showed that the reaction was complete. The mixture was cooled to 20 ℃. The aqueous layer was extracted with toluene and the aqueous phase was extracted with ethyl acetate (200 ml x 3). The combined organic phases were washed with brine (100 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (3% ethyl acetate/petroleum ether) to give 4-bromo-2-methoxy-6-methyl-benzonitrile (12.1 g,53.52mmol,38.55% yield) as a yellow solid.
Step 3:
DIBAL-H (1M, 115mL,5.78 eq.) was added dropwise to a mixture of 4-bromo-2-methoxy-6-methyl-benzonitrile (4.5 g,19.91mmol,1 eq.) in THF (50 mL) at-70℃under N2. The mixture was stirred at 20℃for 10 hours. TLC showed new spots. The residue was poured into HCl (2M) to adjust ph=5-6. The aqueous phase was extracted with ethyl acetate (100 ml x 3). The combined organic phases were washed with brine (100 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether) to give 4-bromo-2-methoxy-6-methyl-benzaldehyde (2.3 g,10.04mmol,50.44% yield) as a yellow solid.
Step 4:
to a suspension of 4-bromo-2-methoxy-6-methyl-benzaldehyde (2.3 g,10.04mmol,1 eq.) in t-BuOH (20 mL) was added sodium chlorite (1.82 g,20.08mmol,2 eq.) and NaH 2 PO 4 (4.82 g,40.16mmol,4 eq.) in H 2 O (20 mL). To this solution was added 2-methyl-2-butene (5.63 g,80.32mmol,8.51mL,8 eq.). The resulting homogeneous solution was stirred at 20℃for 1.5 hours. TLC (petroleum ether: ethyl acetate=10:1, rf=0.63) showed that the reaction was complete. LCMS showed the desired MS. The residue was poured into HCl (2M) to adjust ph=5-6. The aqueous phase was extracted with ethyl acetate (20 ml x 3). The organic phase was then treated with NaHCO 3 Adjust to ph=9, wash the combined organic phases with brine (20 ml x 3), pour the residue into HCl (2M) to adjust ph=5-6. The aqueous phase was extracted with ethyl acetate (20 ml x 3), the combined organic phases were washed with brine (20 ml x 3), and dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 4-bromo-2-methoxy-6-methyl-benzoic acid (1.73 g,7.06mmol,70.31% yield) as a yellow solid.
Step 5:
to a solution of 4-bromo-2-methoxy-6-methyl-benzoic acid (1.73 g,7.06mmol,1 eq.) in DMF (20 mL) was added MeI (2.00 g,14.12mmol,878.92uL,2 eq.) and K2CO3 (2.93 g,21.18mmol,3 eq.). The mixture was stirred at 20℃for 1 hour. TLC showed the reaction was complete. Pouring the residue intoIn water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give methyl 4-bromo-2-methoxy-6-methyl-benzoate (1.17 g,4.52mmol,63.97% yield) as a white solid.
Step 6:
to a stirred solution of methyl 4-bromo-2-methoxy-6-methyl-benzoate (1.17 g,4.52mmol,1 eq.) in DCE (10 mL) under nitrogen was added NBS (964.47 mg,5.42mmol,1.2 eq.) followed by 2- [ (E) - (1-cyano-1-methyl-ethyl) azo]2-methyl-propionitrile (37.08 mg,225.78 mol,0.05 eq) and the resulting mixture was stirred vigorously at 70℃for 2h. LCMS showed the desired MS. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18. Times.40 mm. Times.10 um; mobile phase: [ water (0.225% FA) -ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:55% -85%,10 min) to give methyl 4-bromo-2- (bromomethyl) -6-methoxy-benzoate (550 mg,1.63mmol,36.04% yield) as a white solid.
Step 7:
to a mixture of methyl 4-bromo-2- (bromomethyl) -6-methoxy-benzoate (550 mg,1.63mmol,1 eq.) and 3-aminopiperidine-2, 6-dione (321.40 mg,1.95mmol,1.2 eq., HCl) in DMF (10 mL) was added DIEA (1.05 g,8.14mmol,1.42mL,5 eq.) at 20 ℃ in one portion. The mixture was stirred at 85 ℃ for 48 hours. LCMS showed the desired MS. The crude product was concentrated in vacuo. The crude product was purified by MeCN (20 mL) andH 2 o (20 mL) was ground at 20deg.C. The crude product was filtered and the solid concentrated in vacuo to give 3- (5-bromo-7-methoxy-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (420 mg,1.19mmol,73.08% yield) as a dark grey solid.
Step 8:
to a solution of 3- (5-bromo-7-methoxy-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (200 mg,566.30umol,1 eq.) and 4- (dimethoxymethyl) piperidine (180.34 mg,1.13mmol,2 eq.) in DMSO (5 mL) was added Cs 2 CO 3 (369.03 mg,1.13mmol,2 eq.) and Pd-PEPSI-pent Cl-o-methylpyridine (30.75 mg,56.63umol,0.1 eq.). After addition, the reaction mixture was taken up in N at 80 ℃ 2 Stirring was carried out for 12h. LCMS showed the desired MS. The mixture was cooled to 20 ℃. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by preparative TLC (petroleum ether/ethyl acetate=0/1) to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl as a yellow oil]-7-methoxy-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (268 mg,291.92umol,51.55% yield, 47% purity).
Step 9:
at 20 ℃ at N 2 Downward 3- [5- [4- (dimethoxymethyl) -1-piperidinyl]-7-methoxy-1-oxo-isoindolin-2-yl]To a mixture of piperidine-2, 6-dione (268 mg,621.12umol,1 eq.) in THF (5 mL) was added HCl (2M, 5mL,16.10 eq.) in one portion. The mixture was stirred at 20 ℃ for 1 hour to give a yellow solution. LCMS showed the desired MS. Will remain behindPouring the mixture into saturated NaHCO 3 To adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 1- [2- (2, 6-dioxo-3-piperidinyl) -7-methoxy-1-oxo-isoindolin-5-yl as a yellow solid]Piperidine-4-carbaldehyde (131 mg,237.93umol,38.31% yield, 70% purity).
Step 10:
at 20 ℃ at N 2 Downward 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Pyrimidin-4-yl]-2H-indazole (56.90 mg,127.14umol,1 eq) and 1- [2- (2, 6-dioxo-3-piperidinyl) -7-methoxy-1-oxo-isoindolin-5-yl]To a mixture of piperidine-4-carbaldehyde (70 mg,127.14umol,70% purity, 1 eq.) in MeOH (10 mL) was added borane in one portion; 2-methylpyridine (27.20 mg, 254.27. Mu.l, 2 eq.) and HOAc (7.63 mg, 127.14. Mu.l, 7.27. Mu.l, 1 eq.). The mixture was stirred at 20℃for 10h to give a yellow solution. LCMS showed the desired MS. Pouring the obtained product into H 2 O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -30%,40 min) to give 3- [ 7-methoxy-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] as a white solid ]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (61 mg,73.70umol,57.97% yield, 98.71% purity, FA).
Exemplary synthesis of compound 216: compound 216 was prepared in a similar manner to compound 215.
Exemplary Synthesis of Compound 217
Step 1:
to a dry three-neck round bottom flask was added a solution of diisopropylamine (4.61 g,45.54mmol,6.44mL,2.4 eq.) in THF (45 mL). The flask was cooled to-78 ℃ and n-BuLi solution (2.5 m,17.46ml,2.3 eq.) was added dropwise. The reaction mixture was then stirred at 0℃to 20℃for 30min. A solution of 3, 4-difluorobenzoic acid (3 g,18.98mmol,1 eq.) in THF (30 mL) was added dropwise at-78deg.C, and the mixture was stirred for 1.5 hours, followed by addition of excess carbon dioxide particles, and the mixture was further stirred at-78deg.C for 15min, followed by warming to 20deg.C. The reaction mixture was then stirred at 20℃for 2h. A solution of 1N sodium hydroxide in water (50 mL) was added. The resulting mixture was extracted with tert-butyl methyl ether (20 mL. Times.2). The aqueous layer was acidified to pH 1 with hydrogen chloride (6N) and extracted with ethyl acetate (40 mL. Times.3). The combined ethyl acetate extracts were dried over anhydrous sodium sulfate and concentrated to give 3, 4-difluorophthalic acid (4 g, crude) as a white solid. The crude product was used directly in the next step.
Step 2:
to a solution of 3, 4-difluorophthalic acid (4 g,19.79mmol,1 eq.) and 3-aminopiperidine-2, 6-dione (4.89 g,29.69mmol,1.5 eq., HCl) in HOAc (6 mL) was added NaOAc (4.87 g,59.37mmol,3 eq.). The mixture was stirred at 120℃for 12h. LCMS showed the reaction was complete. The reaction mixture was quenched by the addition of water (200 mL) at 0 ℃ and the solid was collected by filtration. 2- (2, 6-dioxo-3-piperidinyl) -4, 5-difluoro-isoindoline-1, 3-dione (3.3 g,11.22mmol,56.67% yield) was obtained as a black solid. The residue was used in the next step without further purification.
Step 3:
to a solution of 2- (2, 6-dioxo-3-piperidinyl) -4, 5-difluoro-isoindoline-1, 3-dione (1 g,3.40mmol,1 eq.) and tert-butyl 4- (4-piperidinylmethyl) piperazine-1-carboxylate (1.00 g,3.53mmol,1.04 eq.) in DMSO (20 mL) was added DIEA (1.32 g,10.20mmol,1.78mL,3 eq.). After the addition, the reaction mixture was stirred at 60 ℃ for 1h. LCMS showed the reaction was complete. After cooling, the reaction mixture was diluted with ethyl acetate (40 mL) and washed with brine (30 mL x 3). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-1, 3-dioxo-isoindolin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (780 mg,1.40mmol,41.15% yield) as a yellow solid.
Step 4:
to a solution of tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-1, 3-dioxo-isoindolin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (780 mg,1.40mmol,1 eq.) in HOAc (10 mL) was added Zn (3.27 g,50.01mmol,35.75 eq.). After the addition, the reaction mixture was stirred at 90 ℃ for 1h. LCMS showed the reaction was complete. The reaction mixture was filtered, and the filtrate was concentrated. The residue was dissolved in water (30 mL) and adjusted to pH 7-8 with saturated sodium bicarbonate solution. The mixture was extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (30 ml×2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: xtime C18:40 mm 10um; mobile phase: [ water (0.225% FA) -ACN ]; B%:0% -30%,10 min) to give tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-1-oxo-isoindol-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (280 mg,504.76umol,36.08% yield, 98% purity) as a white solid.
Step 5:
to a solution of tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -5-fluoro-1-oxo-isoindolin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (100 mg,183.95umol,1 eq.) in DCM (3 mL) was added TFA (770.00 mg,6.75mmol,0.5mL,36.71 eq.). After the addition, the reaction mixture was stirred at 20 ℃ for 1h. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to give 3- [ 5-fluoro-1-oxo-4- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindolin-2-yl ] piperidine-2, 6-dione (100 mg, crude, TFA) as a pale yellow gum.
Step 6:
to a mixture of 3- [ 5-fluoro-1-oxo-4- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] isoindolin-2-yl ] piperidine-2, 6-dione (100 mg,179.36umol,1 eq, TFA) in MeOH (5 mL) was added DIEA (23.18 mg,179.36umol,31.24ul,1 eq). 1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (70 mg,185.46 mol,1.03 eq) and HOAc (0.5 mL) were then added. Then borane is added; 2-methylpyridine (38.37 mg, 358.72. Mu. Mol,2 eq.) was added to the above solution. After the addition, the reaction solution was stirred at 20 ℃ for 12h. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove most of the solvent. The residue was purified by preparative HPLC (column: xtime C18:40 mm 10um; mobile phase: [ water (FA) -ACN ]; B%:0% -40%,40 min) to afford 3- [ 5-fluoro-4- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (75.1 mg,90.75umol,50.60% yield, 97.27% purity) as a white solid.
Exemplary Synthesis of Compound 218
Step 1:
To a solution of tert-butyl 4- (3-hydroxycyclobutoxy) piperidine-1-carboxylate (2.8 g,10.32mmol,1 eq.) pyridin-4-ol (1.18 g,12.38mmol,1.2 eq.) in tetrahydrofuran (25 mL) was added triphenylphosphine (3.25 g,12.38mmol,1.2 eq.) and diisopropyl azodicarboxylate (2.50 g,12.38mmol,2.4mL,1.2 eq.) under nitrogen at 0 ℃. The reaction mixture was stirred at 50℃for 12h. LCMS showed detection of the desired MS. The mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=10:1 to 0:1). Tert-butyl 4- [3- (4-pyridyloxy) cyclobutoxy ] piperidine-1-carboxylate (3 g,8.61mmol,83% yield) was obtained as a pale yellow solid.
Step 2:
to a solution of tert-butyl 4- [3- (4-pyridyloxy) cyclobutoxy ] piperidine-1-carboxylate (3 g,8.61mmol,1 eq.) in acetonitrile (30 mL) was added benzyl bromide (1.77 g,10.33mmol,1.2mL,1.2 eq.). The reaction mixture was stirred at 80℃for 1h. LCMS showed detection of the desired MS. The mixture was concentrated in vacuo. Tert-butyl 4- [3- [ (1-BLAH-1-benzyl-4-pyridinyl) oxy ] cyclobutoxy ] piperidine-1-carboxylate (4.5 g, crude) was obtained as a yellow oil.
Step 3:
To a solution of tert-butyl 4- [3- [ (1-BLAH-1-benzyl-4-pyridinyl) oxy ] cyclobutoxy ] piperidine-1-carboxylate (4.5 g,8.66mmol,1 eq.) in methanol (40 mL) was added sodium borohydride (819 mg,21.66mmol,2.5 eq.) in three portions at 0deg.C. The reaction mixture was stirred at 20℃for 1h. LCMS showed detection of the desired MS. Hydrochloric acid solution (1M) was added to quench the reaction and water (100 mL) was added to quench the reaction, and the aqueous phase was extracted with ethyl acetate (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether: ethyl acetate=10:1 to 0:1). Tert-butyl 4- [3- [ (1-benzyl-3, 6-dihydro-2H-pyridin-4-yl) oxy ] cyclobutoxy ] piperidine-1-carboxylate (900 mg,1.89mmol,22% yield, 93% purity) was obtained as a pale yellow solid.
Step 4:
to a solution of tert-butyl 4- [3- [ (1-benzyl-3, 6-dihydro-2H-pyridin-4-yl) oxy ] cyclobutoxy ] piperidine-1-carboxylate (900 mg,2.03mmol,1 eq.) in ethanol (50 mL) under nitrogen was added palladium on activated carbon (100 mg,10% purity) and palladium hydroxide on carbon (100 mg,0.14mmol,20% purity, 0.07 eq.). The reaction mixture was stirred at 50℃under hydrogen (50 Psi) for 16h. TLC (petroleum ether: ethyl acetate=1:1) showed that a new spot was detected. LCMS showed detection of the desired MS. The mixture was filtered and the filtrate was concentrated in vacuo. The residue was used in the next step. Tert-butyl 4- [3- (4-piperidinyloxy) cyclobutoxy ] piperidine-1-carboxylate (750 mg, crude) was obtained as a pale yellow oil.
Step 5:
to a solution of 3- (5-bromo-6-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (500 mg,1.47mmol,1 eq), tert-butyl 4- [3- (4-piperidinyloxy) cyclobutoxy ] piperidine-1-carboxylate (627 mg,1.76mmol,1.2 eq) in dimethyl sulfoxide (5 mL) was added cesium carbonate (955 mg,2.93mmol,2 eq) and Pd-PEPPSI-pentcl-o-methylpyridine (142 mg,0.15mmol,0.1 eq) under nitrogen. The reaction was stirred at 80℃for 12h. LCMS showed detection of the desired MS. Dichloromethane (20 mL) and water (20 mL) were added to the mixture and the aqueous phase was extracted with dichloromethane (20 mL x 2). The combined organic phases were washed with brine (20 mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by preparative TLC (dichloromethane: methanol=10:1). 4- [3- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylic acid tert-butyl ester (120 mg,0.19mmol,13% yield) was obtained as a pale yellow solid.
Step 6:
to a solution of tert-butyl 4- [3- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] cyclobutoxy ] piperidine-1-carboxylate (120 mg,0.19mmol,1 eq.) in dichloromethane (5 mL) was added trifluoroacetic acid (1.54 g,13.51mmol,1mL,69.19 eq.). The reaction mixture was stirred at 20℃for 0.5h. LCMS showed detection of the desired MS. The mixture was concentrated in vacuo. The residue was used directly in the next step. 3- [ 4-fluoro-1-oxo-5- [4- [3- (4-piperidinyloxy) cyclobutoxy ] -1-piperidinyl ] isoindolin-2-yl ] piperidine-2, 6-dione (120 mg,0.19mmol,97% yield, trifluoroacetate) was obtained as a yellow oil.
Step 7:
to a solution of 3- [ 4-fluoro-1-oxo-5- [4- [3- (4-piperidinyloxy) cyclobutoxy ] -1-piperidinyl ] isoindolin-2-yl ] piperidine-2, 6-dione (120 mg,0.19mmol,1 eq, trifluoroacetate) in dimethyl sulfoxide (5 mL) was added N, N-diisopropylethylamine (123 mg,0.95mmol,0.2mL,5 eq) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine (104 mg,0.19mmol,1 eq). The reaction mixture was stirred at 90℃for 2h. LCMS showed detection of the desired MS. Water (50 mL) was added to the mixture and the aqueous phase was extracted with ethyl acetate (50 mL x 3). The combined organic phases were washed with brine (50 ml x 2), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by preparative TLC (petroleum ether: ethyl acetate=1:2). 3- [ 4-fluoro-5- [4- [3- [ [1- [6- [5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclobutoxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (120 mg,0.11mmol,57% yield, 93% purity) was obtained as a pale yellow solid.
Step 8:
to a solution of 3- [ 4-fluoro-5- [4- [3- [ [1- [6- [5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclobutoxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (120 mg,0.12mmol,1 eq.) in tetrahydrofuran (2 mL) was added hydrochloric acid (2 m,2mL,34.07 eq.). The reaction mixture was stirred at 40℃for 12h. LCMS showed detection of the desired MS. The mixture was concentrated in vacuo. The residue was purified by preparative HPLC (column: unisil 3-100C18 Ultra150*50mm*3um; mobile phase: [ water (FA) -ACN ]; B%:25% -55%,10 min). 3- [ 4-fluoro-5- [4- [3- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-pyrazolo [3,4-c ] pyridin-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] oxy ] cyclobutoxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (35.4 mg,0.04mmol,38% yield, 98% purity) was obtained as a white solid.
Exemplary Synthesis of Compound 219
Step 1:
to a solution of methyl 3,4, 5-trifluorobenzoate (2.5 g,13.15mmol,1 eq.) and 4- (dimethoxymethyl) piperidine (2.09 g,13.15mmol,1 eq.) in MeCN (15 mL) was added TEA (3.99 g,39.45mmol,5.49mL,3 eq.) at 70℃under N 2 Stirred for 16h. TLC (petroleum ether: ethyl acetate=3:1, rf=0.3) showed a new spot for the reaction. Passing the reactant through NH 4 The Cl (20 mL) solution was quenched and extracted with ethyl acetate (3 x 20 mL). The combined organic phases were washed with water, with Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=50/1, 1/1) to give 4- [4- (dimethoxymethyl) -1-piperidinyl as a white solid]-methyl 3, 5-difluoro-benzoate (3.4 g,7.17mmol,54.56% yield, 69.5% purity).
Step 2:
to 4- [4- (dimethoxymethyl) -1-piperidinyl]-methyl 3, 5-difluoro-benzoate (3.4 g,10.32mmol,1 eq.) in THF (7 mL), meOH (7 mL) and H 2 To a solution of a mixture of O (7 mL) was added NaOH (2.06 g,51.62mmol,5 eq.). The mixture was stirred at 50℃for 2 hours. The mixture was concentrated to remove most of the organic solvent. The residue was extracted with ethyl acetate (30 mL). The pH of the aqueous phase was adjusted to about 4 with 0.5N hydrochloric acid, followed by extraction with ethyl acetate (30 mL. Times.3). The combined organic layers were washed with brine, dried (sodium sulfate) and concentrated. The crude product was used directly in the next step. Obtaining 4- [4- (dimethoxymethyl) -1-piperidine as a white solid Base group]-3, 5-difluoro-benzoic acid (3.2 g, crude).
Step 3:
to a solution of 4- [4- (dimethoxymethyl) -1-piperidinyl ] -3, 5-difluoro-benzoic acid (2.5 g,7.93mmol,1 eq.) in THF (20 mL) was added n-BuLi (2.5 m,9.51mL,3 eq.) dropwise at-78 ℃. After the addition, the mixture was stirred at this temperature for 0.5 h, then DMF (5.80 g,79.29mmol,6.10mL,10 eq.) was added dropwise at-78 ℃. The resulting mixture was stirred at-78 ℃ for 0.5 hours. TLC (petroleum ether: ethyl acetate=3:1) detected a major new spot. The mixture was quenched with saturated ammonium chloride solution, followed by adjusting the pH to about 4 with 0.5N hydrogen chloride. The mixture was partitioned between ethyl acetate (30 mL) and water (20 mL). The aqueous phase was extracted with ethyl acetate (30 mL. Times.2). The combined organic layers were washed with brine, dried (sodium sulfate) and concentrated. The crude material was triturated with petroleum ether/ethyl acetate (40 mL) to give 4- [4- (dimethoxymethyl) -1-piperidinyl ] -3, 5-difluoro-2-formyl-benzoic acid (2.4 g, crude) as a yellow solid.
Step 4:
to a solution of 3-aminopiperidine-2, 6-dione (1.73 g,10.49mmol,1.5 eq., HCl) in MEOH (30 mL) was added NaOAc (2.29 g,27.96mmol,4 eq.). The mixture was stirred at 25℃for 0.5 h. Addition of 4- [4- (dimethoxymethyl) -1-piperidinyl ]-3, 5-difluoro-2-formyl-benzoic acid (2.4 g,6.99mmol,1 eq.) and HOAc (1.68 g,27.96mmol,1.60ml,4 eq.) and the mixture was stirred at 25 ℃ for 0.5 hours. Adding NaBH 3 CN (1.32 g,20.97mmol,3 eq.) and the mixture was stirred at 25℃for 2 hours. The mixture was concentrated under reduced pressure to remove most of the solvent. The mixture was purified in ethyl acetate (20mL) and water (20 mL). The pH of the mixture was adjusted to about 4 with 0.5N hydrochloric acid. The mixture was filtered, the filter cake was collected and dried to give a white solid. The crude product was used directly in the next step. Obtaining 4- [4- (dimethoxymethyl) -1-piperidinyl as a white solid]-2- [ [ (2, 6-dioxo-3-piperidyl) amino group]Methyl group]-3, 5-difluoro-benzoic acid (1.94 g,3.54mmol,50.57% yield, 83% purity).
Step 5:
to a solution of 4- [4- (dimethoxymethyl) -1-piperidinyl ] -2- [ [ (2, 6-dioxo-3-piperidinyl) amino ] methyl ] -3, 5-difluoro-benzoic acid (1.94 g,4.26mmol,1 eq.) in DMF (15 mL) was added HATU (1.94 g,5.11mmol,1.2 eq.) followed by DIEA (1.65 g,12.78mmol,2.23mL,3 eq.) to the mixture which was stirred at 25℃for 2h. The mixture was poured into water (50 mL), the mixture was filtered, and the filter cake was dried under vacuum. The residue was triturated with petroleum ether and ethyl acetate (20 ml, V/v=1/1).
The filter cake was dried under vacuum to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl ] -4, 6-difluoro-1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (1.5 g,3.36mmol,78.89% yield, 98% purity) as a white solid.
Step 6:
to 3- [5- [4- (dimethoxymethyl) -1-piperidinyl]-4, 6-difluoro-1-oxo-isoindolin-2-yl]To a solution of piperidine-2, 6-dione (1 g,2.29mmol,1 eq.) in THF (5 mL) was added HCl (2M, 5mL,4.37 eq.) and at 40℃under N 2 Stirred for 2h. LCMS showed the desired product. Pouring the reaction mixture into H 2 O (20 mL) with NaHCO 3 The aqueous solution was basified until ph=8. The mixture was treated with acetic acidEthyl ester (20 ml x 5) extraction. Anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was used directly in the next step without any purification. The compound 1- [2- (2, 6-dioxo-3-piperidinyl) -4, 6-difluoro-1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (500 mg,1.20mmol,52.53% yield, 94% purity) was found to be a white solid.
Step 7:
to a mixture of 1- [2- (2, 6-dioxo-3-piperidinyl) -4, 6-difluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (50 mg,127.76umol,1 eq.) in HOAC (1 mL) and MeOH (10 mL) was added a solution of 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -1H-indazole (92.76 mg,191.64umol,1.5 eq., HCl) and DIEA (49.53 mg,383.27umol,66.76uL,3 eq.) in MeOH (0.5 mL). Then borane is added; 2-methylpyridine (27.33 mg,255.51umol,2 eq.) was added to the above solution. After the addition, the reaction solution was stirred at 20 ℃ for 16h. LCMS showed the desired product. The residue was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: xtime C18. Times.30 mm. 10um; mobile phase: [ water (FA) -ACN ]; B%:0% -35%,35 min) to afford 3- [4, 6-difluoro-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (42.3 mg,50.89umol,39.83% yield, 99% purity) as a white solid.
Exemplary synthesis of compound 220: compound 220 is prepared in a similar manner to compound 219.
Exemplary synthesis of compound 221: compound 221 was prepared in an analogous manner to compound 181 using (3S) -3-methylpiperazine-1-carboxylic acid tert-butyl ester.
Exemplary synthesis of compound 222: compound 221 was prepared in an analogous manner to compound 181 using (2S) -2-methylpiperazine-1-carboxylic acid tert-butyl ester.
Exemplary synthesis of compound 223: compound 223 is prepared in a similar manner to compound 219 using 4-fluoro-4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperidine-1-carboxylic acid tert-butyl ester.
Step 1:
at 70 ℃ at N 2 To a mixture of 4-methylpyridine (5 g,53.69mmol,5.26mL,1 eq.) in THF (100 mL) was added n-BuLi (2.5M, 25.77mL,1.2 eq.) dropwise. The mixture was stirred at-70℃for 3h to give an orange solution, followed by dropwise addition of tert-butyl 4-oxopiperidine-1-carboxylate (10.70 g,53.69mmol,1 eq.) in THF (10 mL) and stirring of the solution at 20℃for 2 h to give a pale yellow solution. LCMS showed formation of the desired compound. The mixture was poured into water (50 mL). The aqueous phase was extracted with ethyl acetate (40 ml x 3). The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=0/1) to give tert-butyl 4-hydroxy-4- (4-pyridylmethyl) piperidine-1-carboxylate (10.33 g,35.33mmol,65.81% yield) as a pale yellow liquid.
Step 2:
at-60 ℃ at N 2 To a mixture of tert-butyl 4-hydroxy-4- (4-pyridylmethyl) piperidine-1-carboxylate (10 g,34.20mmol,1 eq.) in DCM (100 mL) was added dropwise N-ethyl-N- (trifluoro- λ4-sulfanyl) ethylamine (6.62 g,41.04mmol,5.42mL,1.2 eq.). The mixture was stirred at-60℃for 10 hours. TLC (petroleum ether: ethyl acetate=0/1) showed the reactionHas been completed. Cool the reaction to 0deg.C and use NaHCO 3 The aqueous solution was quenched to adjust ph=7-8. The aqueous phase was extracted with DCM (60 ml x 2). The combined organic layers were washed with brine (20 ml x 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1/1) to give tert-butyl 4-fluoro-4- (4-pyridylmethyl) piperidine-1-carboxylate (7.45 g, crude) as a colorless oil.
Step 3:
to a solution of tert-butyl 4-fluoro-4- (4-pyridylmethyl) piperidine-1-carboxylate (2 g,6.79mmol,1 eq.) in EtOH (20 mL) and HOAc (408.01 mg,6.79mmol,388.58uL,1 eq.) was added PtO2 (231.43 mg,1.02mmol,0.15 eq.) at 25 ℃. The mixture is then subjected to H at 70 DEG C 2 Stirring was carried out at (50 psi) for 24h. TLC (petroleum ether: ethyl acetate=3/1) showed the reaction was complete. The suspension was filtered through a pad of celite and the pad or filter cake was washed with EtOH (100 mL x 3). The combined filtrates were concentrated to dryness to give the product. The residue was used in the next step without further purification. The product, tert-butyl 4-fluoro-4- (4-piperidinylmethyl) piperidine-1-carboxylate (2.7 g, crude, HOAC), was a brown solid.
Step 4:
at 90 ℃ at N 2 Downward 4-fluoro-4- (4-piperidylmethyl) piperidine-1-carboxylic acid tert-butyl ester (200 mg,554.84umol,1 eq., HOAC) and 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl]Methoxy group]To a mixture of ethyl-trimethyl-silane (239.14 mg,554.84umol,1 eq.) in DMSO (4 mL) was added DIEA (358.55 mg,2.77mmol,483.22uL,5 eq.) in one portion. The mixture was stirred at 90℃for 2 hours. LCMS showed the reaction was completeAnd (3) forming the finished product. The mixture was cooled to 25 ℃ and poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 2). The combined organic phases were washed with brine (5 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was used in the next step without further purification. The product 4-fluoro-4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] was obtained as a pale yellow oil ]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Tert-butyl piperidine-1-carboxylate (45 mg,60.87umol,16.27% yield, 94% purity).
Exemplary Synthesis of Compound 224
Step 1:
to a solution of benzyl 4-formylpiperidine-1-carboxylate (1.78 g,7.21mmol,1 eq.) and HOAC (25.98 mg,432.69umol,24.75uL,0.06 eq.) in DCM (10 mL) was added tert-butyl 3, 3-dimethylpiperazine-1-carboxylate (1.70 g,7.93mmol,1.1 eq.) followed by the mixture at 25℃under N 2 Stir for 20 hours, then stir NaBH (OAc) 3 (2.29 g,10.80mmol,1.50 eq.) was added to the above mixture and the mixture was stirred at 25℃for 2 hours. By addition of saturated NaHCO 3 Adjustment to ph=8-9 the reaction mixture was quenched and extracted with DCM (20 ml x 3). The combined organic layers were washed with brine (50 ml x 2), dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel (0% -25% ethyl acetate/petroleum) to give tert-butyl 4- ((1- ((benzyloxy) carbonyl) piperidin-4-yl) methyl) -3, 3-dimethylpiperazine-1-carboxylate (2.11 g,4.73mmol,65.57% yield) as a colorless oil.
Step 2:
at N 2 Downward 4- [ (1-benzyloxycarbonyl-4-piperidinyl) methyl group ]To a solution of tert-butyl 3, 3-dimethyl-piperazine-1-carboxylate (2.1 g,4.71mmol,1 eq.) in MeOH (20 mL) was added Pd/C (500 mg,10% purity). The suspension was degassed under vacuum and treated with H 2 Purging several times. The mixture was stirred at 25℃under H 2 Stirring for 1 hour at (15 psi). TLC (petroleum ether: ethyl acetate=1:1) showed the reaction was complete. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 3, 3-dimethyl-4- (4-piperidylmethyl) piperazine-1-carboxylate (1.39 g,4.46mmol,94.70% yield) as a white gum.
Step 3:
to 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl]Methoxy group]To a solution of ethyl-trimethyl-silane (0.15 g,348.03umol,1 eq.) in DMSO (3 mL) were added TEA (176.08 mg,1.74mmol,242.20uL,5 eq.) and tert-butyl 3, 3-dimethyl-4- (4-piperidylmethyl) piperazine-1-carboxylate (216.79 mg,696.05umol,2 eq.). The mixture was stirred at 100℃for 1 hour. TLC indicated 2- [ [3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) indazol-2-yl]Methoxy group]The ethyl-trimethyl-silane was completely consumed and a new spot formed. The reaction mixture was quenched by the addition of EtOAc (50 mL) in water 50mL at 25 ℃. The organic layer was washed with water (30 ml x 3), dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel4g/>Silica gel flash column, eluent: 0% -20% ethyl acetate/petroleum ether gradient, 50 mL/min) to give 3, 3-dimethyl-4- [ [1- [6- [5- (1-methylcyclopropoxy) ] a pale yellow solid2- (2-trimethylsilylethoxymethyl) indazol-3-yl]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]Tert-butyl piperazine-1-carboxylate (0.19 g,266.99umol,76.72% yield, 99.210% purity).
Step 4:
to a solution of tert-butyl 3, 3-dimethyl-4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylate (0.18 g,254.96umol,1 eq.) in MeOH (3 mL) was added HCl/EtOAc (4 m,127.48ul,2 eq.). The mixture was stirred at 25℃for 4 hours. LC-MS (EB 4455-23-P1B) showed that 3, 3-dimethyl-4- [ [1- [6- [5- (1-methylcyclopropoxy) -2- (2-trimethylsilylethoxymethyl) indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazine-1-carboxylic acid tert-butyl ester was completely consumed and one major peak with the desired mass was detected. The mixture was concentrated under reduced pressure. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 3- [6- [4- [ (2, 2-dimethylpiperazin-1-yl) methyl ] -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1H-indazole (0.13 g, crude, HCl) as a white solid.
Step 5:
to a solution 1 of 3- [6- [4- [ (2, 2-dimethylpiperazin-1-yl) methyl ] -1-piperidinyl ] pyrimidin-4-yl ] -5- (1-methylcyclopropoxy) -1H-indazole (0.13 g,253.86umol,1.58 eq, HCl) in MeOH (5 mL) was added DIEA (60 mg,464.24umol,80.86uL,2.89 eq). The mixture was stirred at 25℃for 60min. To a solution 2 of 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (60 mg,160.70umol,1 eq.) in MeOH (2 mL) was then added AcOH (9.65 mg,160.70umol,9.19uL,1 eq.). Solution 2 was stirred at 25℃for 60min. Then adding solution 1 and borane to solution 2; 2-methylpyridine (34.38 mg,321.40 mol,2 eq.) then the mixture was stirred under stirring for 16 hours. LCMS showed complete consumption of 1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde and detection of the desired mass. The residue was purified by preparative HPLC (column: gemini NX C18. Times.10X 150mm; mobile phase: [ water (FA) -ACN ]; B%:10% -40%,20 min) to give 3- [5- [4- [ [3, 3-dimethyl-4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -4-fluoro-1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (57.4 mg,65.84 mol,40.97% yield, 99.737% purity) as a white solid.
Exemplary synthesis of compound 225: compound 225 was prepared in a similar manner to compound 224 starting from tert-butyl 3, 6-diazabicyclo [3.1.1] heptane-6-carboxylate.
Exemplary synthesis of compound 226: compound 226 is prepared in a similar manner to compound 224 starting from tert-butyl (1S, 4S) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate.
Exemplary synthesis of compound 227: compound 227 was prepared in a similar manner to compound 210.
Exemplary Synthesis of Compound 228
Step 1:
to a solution of methyl 2-bromo-3, 4-difluoro-benzoate (1.2 g,4.78mmol,1.52 eq.) in MeCN (10 mL) and DMSO (3 mL) at 20deg.C was added TEA (1.22 g,12.02mmol,1.67mL,3.83 eq.) and 4- [4- (4-piperidinyloxy) cyclohexyloxy]Tert-butyl piperidine-1-carboxylate (1.2 g,3.14mmol,1 eq.) and the reaction mixture was stirred at 70℃for 10 hours. TLC showed new spots. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -15% ethyl acetate/petroleum ether, 15% ethyl acetate/petroleum ether) to give 4- [4- [ [1- (3-bromo-2-fluoro-4-methoxycarbonyl-phenyl) -4-piperidinyl ] as a white solid ]Oxy group]Cyclohexyloxy radical]Tert-butyl piperidine-1-carboxylate (700 mg,1.09mmol,34.86% yield, 95.84% purity).
Step 2:
to 4- [4- [ [1- (3-bromo-2-fluoro-4-methoxycarbonyl-phenyl) -4-piperidinyl]Oxy group]Cyclohexyloxy radical]To a solution of tert-butyl piperidine-1-carboxylate (500 mg, 814.92. Mu. Mol,1 eq.) in DMF (2 mL) was added tert-butyl isocyanate (135.49 mg,1.63mmol, 184.09. Mu.L, 2 eq.) Pd (OAc) 2 (7.32 mg,32.60umol,0.04 eq.) PCy 3 (9.14 mg,32.60umol,10.57uL,0.04 eq.) Na 2 CO 3 (86.37 mg,814.92umol,1 eq.) and Et 3 SiH (284.28 mg,2.44mmol,390.49uL,3 eq.). The mixture was stirred in a polytetrafluoroethylene tank (teflon tank) at 100 ℃ for 24 hours. LCMS showed the desired MS. The mixture was cooled to 20 ℃. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -18% ethyl acetate/petroleum ether, 18% ethyl acetate/petroleum ether) to give 4- [4- [ [1- (2-fluoro-3-formyl-4-methoxycarbonyl-phenyl) -4-piperidinyl ] as a yellow solid ]Oxy group]Cyclohexyloxy radical]Tert-butyl piperidine-1-carboxylate (400 mg,603.55umol,18.52% yield, 84.9% purity).
Step 3:
at 25 ℃ at N 2 Downward 4- [4- [ [1- (2-fluoro-3-formyl-4-methoxycarbonyl-phenyl) -4-piperidinyl]Oxy group]Cyclohexyloxy radical]To a mixture of tert-butyl piperidine-1-carboxylate (300 mg,533.17 mol,1 eq) and 3-aminopiperidine-2, 6-dione (131.63 mg,799.76 mol,1.5 eq, HCl) in MeOH (10 mL) was added HOAc (0.5 mL) and borane in one portion; 2-methylpyridine (114.06 mg,1.07mmol,2 eq.). The mixture was stirred at 25 ℃ for 16 hours to give a red suspension. LCMS showed the desired MS. The reaction mixture was filtered and the filter cake was washed with 20mL TBME and dried in vacuo to give 4- [4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a red solid]-4-piperidinyl]Oxy group]Cyclohexyloxy radical]Tert-butyl piperidine-1-carboxylate (170 mg,227.46umol,42.66% yield, 86% purity).
Step 4:
at 25 ℃ at N 2 Downward 4- [4- [ [1- [2- (2, 6-dioxo-3-piperidyl) -4-fluoro-1-oxo-isoindolin-5-yl ]]-4-piperidinyl]Oxy group]Cyclohexyloxy radical]To a mixture of tert-butyl piperidine-1-carboxylate (170 mg,264.49umol,1 eq) in DCM (5 mL) was added TFA (3.08 g,27.01mmol,2mL,102.13 eq) in one portion. The mixture was stirred at 25℃for 30min. TLC showed the reaction was complete. The mixture was concentrated in vacuo to give 3- [ 4-fluoro-1-oxo-5- [4- [4- (4-piperidinyloxy) cyclohexyloxy) as a red oil ]-1-piperidinyl group]Isoindolin-2-yl]Piperidine-2, 6-dione (170 mg,199.34umol,75.37% yield, 77% purity, TFA).
Step 5:
to 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (70 mg,232.76umol,8.99e-1 eq.) and 3- [ 4-fluoro-1-oxySubstituted-5- [4- [4- (4-piperidinyloxy) cyclohexyloxy]-1-piperidinyl group]Isoindolin-2-yl]A solution of piperidine-2, 6-dione (170 mg,258.88umol,1 eq., TFA) in DMSO (10 mL) and DIEA (334.58 mg,2.59mmol,450.92uL,10 eq.). The mixture was stirred at 80℃for 10h. LCMS showed the desired product MS. Pouring the obtained product into H 2 O (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (15 ml x 2), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ Water (FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:10% -50%,35 min) to give 3- [ 4-fluoro-5- [4- [4- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] as a red solid]Pyrimidin-4-yl]-4-piperidinyl]Oxy group]Cyclohexyloxy radical]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (62.6 mg,72.92umol,28.17% yield, 94% purity).
Exemplary synthesis of compound 229: compound 229 was prepared in a similar manner to compound 181.
Exemplary synthesis of compound 230: compound 230 was prepared in an analogous manner to compound 224 starting from tert-butyl (1 r,4 r) -5- (4-piperidinylmethyl) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate.
Exemplary Synthesis of Compound 231
Step 1:
to a mixture of 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (600 mg,1.76mmol,1 eq.) and tert-butyl 4-fluoro-4- (4-piperidinylmethyl) piperidine-1-carboxylate (792.57 mg,2.64mmol,1.5 eq.) in DMSO (10 mL) was added Cs 2 CO 3 (1.72 g,5.28mmol,3 eq.) Pd-PEPSI-pent Cl-O-methylpyridine (95.51 mg,175.88umol,0.1 eq.) then the mixture was stirred at 90℃under N 2 Stirring is carried out for 12 hours under an atmosphere. TLC (petroleum ether: ethyl acetate=0:1) showed a new major groupSpots. LCMS showed the desired MS. The resulting product was poured into aqueous FA solution to adjust ph=5-6. The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (20 mL), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) to give 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a yellow solid ]-4-piperidinyl]Methyl group]-4-fluoro-piperidine-1-carboxylic acid tert-butyl ester (700 mg,898.99umol,51.11% yield, 72% purity).
Step 2:
to a mixture of tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -4-fluoro-piperidine-1-carboxylate (700 mg,1.25mmol,1 eq.) in DCM (5 mL) was added TFA (3.08 g,27.01mmol,2mL,21.63 eq.) and the mixture was stirred at 25 ℃ under an atmosphere of N2 for 1 hour. TLC (petroleum ether: ethyl acetate=0:1) showed a new major spot. The resulting product was concentrated in vacuo to give 3- [ 4-fluoro-5- [4- [ (4-fluoro-4-piperidinyl) methyl ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (600 mg,1.09mmol,87.65% yield, 84% purity) as a yellow solid.
Step 3:
to 1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl]Pyrimidin-4-yl]Piperidine-4-carbaldehyde (70 mg,185.46 mol,1 eq.) and 3- [ 4-fluoro-5- [4- [ (4-fluoro-4-piperidinyl) methyl]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (85.41 mg,185.46umol,1 eq.) was added borane to a mixture of MeOH (10 mL) and AcOH (2 mL); 2-methylpyridine (39.67 mg,370.92umol, 2.) The amount) and then the mixture was stirred at 25 ℃ for 12 hours. LCMS showed the desired MS. Pouring the obtained product into H 2 O (10 mL). The mixture was extracted with ethyl acetate (50 ml x 3). The organic phase was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by preparative HPLC (column: xtime C18X 30mm X10 um; mobile phase: [ water (0.225% FA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,40 min) to give 3- [ 4-fluoro-5- [4- [ [ 4-fluoro-1- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl) as a white solid]Pyrimidin-4-yl]-4-piperidinyl]Methyl group]-4-piperidinyl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (81 mg,97.56umol,52.60% yield, 99% purity).
Exemplary synthesis of compound 232: compound 232 was prepared in a similar manner to compound 231.
Exemplary synthesis of compound 233: compound 233 was prepared in a similar manner to compound 126 using the intermediate tert-butyl 4- [3- (4-pyridyloxy) cyclobutoxy ] piperidine-1-carboxylate.
Step 1:
to a mixture of sodium tert-butoxide (3.00 g,31.22mmol,2.82 eq.) and tert-butyl 4- (3-hydroxycyclobutoxy) piperidine-1-carboxylate (3.00 g,11.06mmol,1 eq.) in dimethyl sulfoxide (30 mL) was added 4-chloropyridine (2.16 g,14.37mmol,1.3 eq. Hydrochloride) at 40℃followed by stirring the mixture at 80℃for 12h. LCMS showed the quality of the desired product. To the reaction mixture was added water (50 mL) and extracted with ethyl acetate (30 mL x 3). The combined organic layers were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel 12g/>Silica gel flash column, eluent: 0% -80% ethyl acetate/petroleum ether gradient, 40 mL/min). The compound 4- [3- (4-pyridyloxy) cyclobutoxy was obtained as a yellow solid]Tert-butyl piperidine-1-carboxylate (2.60 g,7.46mmol,67% yield).
Exemplary Synthesis of Compound 234
Step 1:
to a solution of ethyl 4-hydroxycyclohexane formate (20 g,116.13mmol,1 eq.) in THF (200 mL) at 0deg.C were added TMSCl (13.88 g,127.74mmol,16.21mL,1.1 eq.) and TEA (14.10 g,139.36mmol,19.40mL,1.2 eq.) and the reaction mixture was stirred at 25deg.C for 1 hour. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. Then at-65 ℃ at N 2 Et is added dropwise to a stirred solution of the above residue and benzyl 4-oxopiperidine-1-carboxylate (31.15 g,133.55mmol,26.63mL,1.15 eq.) in DCM (300 mL) 3 SiH (20.25 g,174.19mmol,27.82mL,1.5 eq.) and TMSOTF (14.20 g,63.87mmol,11.54mL,0.55 eq.) and the reaction mixture was taken up in N at 0deg.C 2 Stirred for 3 hours. TLC (Petroleum ether: ethyl acetate=3:1, PMA, R) f =0.43) shows that a new spot is formed. The reaction mixture was quenched by the addition of water (300 mL) and extracted with DCM (200 ml×3). The combined organic layers were washed with brine (300 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel120g/>Silica gel flash column, eluent: gradient of 0% -20% ethyl acetate/petroleum ether, 100mL/min). The compound benzyl 4- (4-ethoxycarbonylcyclohexyloxy) piperidine-1-carboxylate (40.8 g,104.75mmol,90.20% yield) was obtained as a colourless oil.
Step 2:
to a solution of benzyl 4- (4-ethoxycarbonylcyclohexyloxy) piperidine-1-carboxylate (40.8 g,104.75mmol,1 eq.) in EtOH (40 mL) was added Pd/C (8 g,104.75mmol,10% purity, 1 eq.) followed by 25℃under H 2 Stirring for 4 hours at (15 psi). TLC (petroleum ether: ethyl acetate=3:1, i 2 ) Showing the formation of new spots. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound ethyl 4- (4-piperidinyloxy) cyclohexanecarboxylate (26.7 g,104.56mmol,99.82% yield) was obtained as a colorless solid.
Step 3:
to a solution of ethyl 4- (4-piperidinyloxy) cyclohexanecarboxylate (10 g,39.16mmol,1 eq.) in THF (70 mL) at 0deg.C was added LiAlH 4 (2.23 g,58.74mmol,1.5 eq.) then the solution was taken up in N at 0deg.C 2 Stirred for 2 hours. TLC (dichloromethane: methanol=5:1) showed complete consumption of starting material. The reaction mixture was quenched by the addition of water (2 mL) and 15% NaOH (4 mL) at 0deg.C, followed by Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. Obtaining the compound [4- (4-piperidinyloxy) cyclohexyl ] as a pale yellow solid]Methanol (7.9 g,37.03mmol,94.57% yield).
Step 4:
to 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (300 mg,879.42umol,1 eq) and [4- (4-piperidinyloxy) cyclohexyl]To a solution of methanol (225.11 mg,1.06mmol,1.2 eq.) in DMSO (10 mL) was added Cs 2 CO 3 (859.60 mg,2.64mmol,3 eq.) and Pd-PEPSI-pent Cl-o-methylpyridine (47.75 mg,87.94umol,0.1 eq.) at 90℃under N 2 Stirred for 16h. LCMS showed about 43% of the desired compound and TLC (petroleum ether: ethyl acetate=0:1, i 2 ) The main spot is shown to be formed. The reaction mixture was quenched by the addition of formic acid (3 mL) at 0deg.C and extracted with EtOAc (50 mL. Times.3). The combined organic layers were washed with brine (30 mL. Times.5), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel12g/>Silica gel flash column, eluent: 0% -100% ethyl acetate/petroleum ether gradient, 60 mL/min). Obtaining the compound 3- [ 4-fluoro-5- [4- [4- (hydroxymethyl) cyclohexyloxy) as a pale yellow solid ]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (95 mg,193.60umol,22.01% yield, 96.5% purity).
Step 5:
to 3- [ 4-fluoro-5- [4- [4- (hydroxymethyl) cyclohexyloxy)]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]To a solution of piperidine-2, 6-dione (95 mg, 200.62. Mu. Mol,1 eq.) in DCM (10 mL) was added DMP (255.27 mg, 601.86. Mu. Mol, 186.33. Mu.L, 3 eq.) and the mixture was stirred at 25℃for 2 hours. TLC (petroleum ether: ethyl acetate=0:1, i 2 ) Showing the formation of new spots. Passing the reaction mixture through a catalyst at 0 DEG CSaturated NaHCO is added at the bottom 3 The pH was adjusted to about 8 for quenching and extracted with DCM (30 mL. Times.3). The combined organic layers were taken up in saturated Na 2 SO 3 (30 ml x 3) and brine (30 ml x 3), washed with Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. Obtaining the compound 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a pale yellow solid]-4-piperidinyl]Oxy group]Cyclohexane carbaldehyde (90 mg,164.15umol,81.82% yield, 86% purity).
Step 6:
to a solution of 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] cyclohexanecarboaldehyde (90 mg,190.87umol,1 eq.) and 5- (1-methylcyclopropoxy) -3- (6-piperazin-1-ylpyrimidin-4-yl) -2H-indazole (97.51 mg,209.96umol,1.1 eq., TFA) in MeOH (20 mL) were added AcOH (2.29 mg,38.17umol,2.18ul,0.2 eq.) and borane; 2-methylpyridine (40.83 mg, 381.74. Mu. Mol,2 eq.) then the solution was stirred at 25℃for 16 hours. LCMS showed about 78% of the desired compound and the starting material was completely consumed. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: xtimate C18 x 30mm x 10um; mobile phase: [ water (FA) -ACN ];: B%:10% -40%,35 min). The compound 3- [ 4-fluoro-5- [4- [4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] cyclohexoxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (40.6 mg,50.04umol,26.22% yield, 99.329% purity) was obtained as a white solid.
Exemplary Synthesis of Compound 235
Step 1:
to [4- (4-piperidinyloxy) cyclohexyl group]To a solution of methanol (3.9 g,15.61mmol,1 eq, HCl) and methyl 2-bromo-4, 5-difluoro-benzoate (3.92 g,15.61mmol,1 eq) in MeCN (10 mL) was added TEA (4.74 g,46.84mmol,6.52mL,3 eq) and the mixture was stirred at 70℃for 16 h. TLC (petroleum ether: ethyl acetate=2:1, uv=254 nm) showed the formation of new spots. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel20g/>Silica gel flash column, eluent: 0% -34% ethyl acetate/petroleum ether gradient, 60 mL/min). Obtaining the compound 2-bromo-5-fluoro-4- [4- [4- (hydroxymethyl) cyclohexyloxy) as a white solid]-1-piperidinyl group]Methyl benzoate (2.5 g,5.63mmol,36.03% yield).
Step 2:
to 2-bromo-5-fluoro-4- [4- [4- (hydroxymethyl) cyclohexyloxy]-1-piperidinyl group]To a solution of methyl benzoate (2.5 g,5.63mmol,1 eq.) in DMF (20 mL) was added tert-butyl isocyanate (935.46 mg,11.25mmol,1.27mL,2 eq.) Pd (OAc) 2 (50.53 mg, 225.06. Mu.mol, 0.04 eq.), PCy3 (78.89 mg, 281.32. Mu.mol, 91.20. Mu.L, 0.05 eq.), na 2 CO 3 (596.34 mg,5.63mmol,1 eq.) and Et 3 SiH (1.96 g,16.88mmol,2.70mL,3 eq.). The mixture was stirred at 65℃under N 2 Stirred for 16 hours. LCMS showed about 37% of the desired compound and TLC (petroleum ether: ethyl acetate=3:1, uv=254 nm) showed formation of the main spot. To the reaction mixture was added water (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layers were washed with brine (50 ml x 5), dried over Na 2 SO 4 Drying, filtering and concentrating under reduced pressure,a residue was obtained. The residue was purified by flash chromatography on silica gel12g/>Silica gel flash column, eluent: 0% -8% ethyl acetate/petroleum ether gradient, 60 mL/min). The compound 5-fluoro-2-formyl-4- [4- [4- (hydroxymethyl) cyclohexyloxy) was obtained as a pale yellow oil]-1-piperidinyl group]Methyl benzoate (1.5 g,3.32mmol,58.95% yield, 87% purity).
Step 3:
to a solution of 3-aminopiperidine-2, 6-dione (585.66 mg,3.56mmol,2 equivalents, HCl) in DCM (15 mL) was added NaOAc (583.80 mg,7.12mmol,4 equivalents), followed by stirring the solution at 25℃for 1 hour, followed by methyl 5-fluoro-2-formyl-4- (4- (((1 r,4 r) -4- (hydroxymethyl) cyclohexyl) oxy) piperidin-1-yl) benzoate (0.7 g,1.78mmol,1 equivalents) and HOAc (21.37 mg,355.83umol,20.35uL,0.2 equivalents) were added to the solution, followed by stirring the solution at 25℃for 0.5 hours, followed by NaBH 3 CN (223.61 mg,3.56mmol,2 eq.) was added to the solution, and the mixture was stirred at 25℃for 16 hours. LCMS showed about 33% compound and TLC (petroleum ether: ethyl acetate=0:1, i 2 ) Indicating complete consumption of starting material. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound 3- (6-fluoro-5- (4- (((1 r,4 r) -4- (hydroxymethyl) cyclohexyl) oxy) piperidin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (250 mg,475.15umol,26.71% yield, 90% purity) was obtained as a white solid.
Step 4:
to a solution of 3- [ 6-fluoro-5- [4- [4- (hydroxymethyl) cyclohexyloxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (400 mg,844.71umol,1 eq.) in DCM (10 mL) was added DMP (1.07 g,2.53mmol,784.55uL,3 eq.) and the solution was stirred at 0deg.C for 2 h. TLC (petroleum ether: ethyl acetate=0:1, pma) showed that new spots formed. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] cyclohexanecarboaldehyde (300 mg, crude) was obtained as a white solid.
Step 5:
to a solution of 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -6-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] cyclohexanecarboaldehyde (100 mg,212.08umol,1 eq.) and 5- (1-methylcyclopropoxy) -3- (6-piperazin-1-ylpyrimidin-4-yl) -2H-indazole (98.50 mg,254.50umol,1.2 eq., HCl) in MeOH (5 mL) was added borane; 2-methylpyridine (45.37 mg, 424.16. Mu. Mol,2 eq.) then the solution was stirred at 25℃for 16 hours. LCMS showed about 45.9% of the desired compound and the starting material was completely consumed. The residue was purified by preparative HPLC (column: xtimate C18 x 30mm x 10um; mobile phase: [ water (FA) -ACN ];: B%:10% -40%,35 min). The compound 3- [ 6-fluoro-5- [4- [4- [ [4- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] cyclohexoxy ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (99.2 mg,115.93umol,54.66% yield, 99.566% purity, FA) was obtained as a white solid.
Exemplary Synthesis of Compound 236
Step 1:
at N 2 Downward 4- [ (1-benzyloxycarbonyl-4-piperidinyl) methyl group]To a solution of tert-butyl 3, 3-dimethyl-piperazine-1-carboxylate (2.1 g,4.71mmol,1 eq.) in MeOH (20 mL) was added Pd/C (500 mg,10% purity). The suspension was degassed under vacuum and treated with H 2 Purging several times. The mixture was stirred at 25℃under H 2 Stirring for 1 hour at (15 psi). TLC (petroleum ether: ethyl acetate=1:1) showed the reaction was complete. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 3, 3-dimethyl-4- (4-piperidylmethyl) piperazine-1-carboxylate (1.39 g,4.46mmol,94.70% yield) as a white gum.
Step 2:
3, 3-dimethyl-4- (4-piperidinylmethyl) piperazine-1-carboxylic acid tert-butyl ester (547.81 mg,1.76mmol,1.5 eq.), 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (400 mg,1.17mmol,1 eq.), pd-PEPSI-pent Cl-o-methylpyridine (31.84 mg,58.63umol,0.05 eq.), cs 2 CO 3 (1.15 g,3.52mmol,3 eq.) A mixture in DMSO (5 mL) is degassed and N 2 Purge 3 times, then stir the mixture at 90 ℃ for 20 hours. LC-MS (EB 4455-29-P1D) showed that 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione was completely consumed and the required mass was detected. The reaction mixture was quenched by addition of HCOOH to ph=2-3 and EtOAc (50 ml x 2) was added. The combined organic layers were washed with brine (50 ml x 2), dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure. And the aqueous layer was extracted with DCM (30 ml x 6). The organic layer was purified by Na 2 SO 4 Dried, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (column height: 4g,100-200 mesh silica gel, 60% ethyl acetate/petroleum ether) to give 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a white solid]-4-piperidinesBase group]Methyl group]-3, 3-dimethyl-piperazine-1-carboxylic acid tert-butyl ester (0.18 g,275.04umol,23.46% yield).
Step 3:
to a solution of tert-butyl 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -3, 3-dimethyl-piperazine-1-carboxylate (170 mg,297.37umol,1 eq.) in DCM (5 mL) was added TFA (1.54 g,13.51mmol,1mL,45.42 eq.). The mixture was stirred at 25℃for 4 hours. TLC showed complete consumption of 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] methyl ] -3, 3-dimethyl-piperazine-1-carboxylic acid tert-butyl ester and detection of the desired spot. The reaction mixture was concentrated under reduced pressure to remove the solvent to give 3- (5- (4- ((2, 2-dimethylpiperazin-1-yl) methyl) piperidin-1-yl) -4-fluoro-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (0.14 g,296.88umol,99.84% yield) as a white solid.
Step 4:
to a solution 1 of 3- [5- [4- [ (2, 2-dimethylpiperazin-1-yl) methyl ] -1-piperidinyl ] -4-fluoro-1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (140 mg,296.88umol,1.60 eq.) in MeOH (5 mL) was added DIEA (70.00 mg,541.62umol,94.34ul,2.92 eq.). The mixture was stirred at 25℃for 20min. To a solution 2 of 1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde (70 mg,185.46 mol,1 eq.) in MeOH (5 mL) was then added AcOH (525.00 mg,8.74mmol,0.5mL,47.14 eq.). Solution 2 was stirred at 25℃for 10min. Then adding solution 1 and borane to solution 2; 2-methylpyridine (39.67 mg,370.92umol,2 eq.) the mixture was then stirred under stirring for 16 hours. LC-MS showed that 1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] piperidine-4-carbaldehyde was completely consumed and the desired quality was detected. The reaction mixture was concentrated under reduced pressure to remove the solvent. The residue was purified by preparative HPLC (column: WHELK-O1 (250 mm. Times. 30mm,5 um); mobile phase: [ water (FA) -ACN ]; B%:10% -50%,35 min) to give 3- [5- [4- [ [2, 2-dimethyl-4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -4-fluoro-1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (88 mg,104.97umol,56.60% yield, 99.360% purity) as a white solid.
Exemplary synthesis of compound 237: compound 237 was prepared in a similar manner to compound 224 starting from tert-butyl 3, 6-diazabicyclo [3.1.1] heptane-6-carboxylate.
Exemplary synthesis of compound 238: compound 238 was prepared in a similar manner to compound 234.
Exemplary Synthesis of Compound 239
Step 1:
to a solution of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (500 mg,1.55mmol,1 eq.) and 4- (dimethoxymethyl) piperidine (369.56 mg,2.32mmol,1.5 eq.) in DMSO (5 mL) was added Cs 2 CO 3 (1.01 g,3.09mmol,2 eq.) and Pd-PEPSI-pent Cl-o-methylpyridine (84.02 mg,154.73umol,0.1 eq.). The mixture was stirred at 80℃for 12 hours. LC-MS (EB 648-394-P1A) showed complete consumption of reactant 1 and detection of a major peak with the desired mass. After cooling, the solution was poured into HCOOH (2 mL) to adjust the pH<7, the reaction solution was diluted with ethyl acetate (30 mL) and washed with water (20 mL. Times.3). The combined organic layers were washed with brine (30 mL. Times.2), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel20g/>Silica gel flash column, eluent: 0% -97% ethyl acetate/petroleum ether gradient, 45 mL/min) to give 3- [5- [4- (dimethoxymethyl) -1-piperidinyl ] as a white solid ]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (315 mg,784.65umol,50.71% yield).
Step 2:
3- [5- [4- (dimethoxymethyl) -1-piperidinyl]-1-oxo-isoindolin-2-yl]A mixture of piperidine-2, 6-dione (310 mg,772.19umol,1 eq), NCS (123.74 mg,926.63umol,1.2 eq), TFA (150.56 mg,1.32mmol,97.77uL,1.71 eq) in DCM (20 mL) and MeOH (2 mL) was degassed and N 2 Purging 3 times, followed by 25℃of the mixture under N 2 Stirring is carried out for 12 hours under an atmosphere. LC-MS (EB 648-396-P1A) showed complete consumption of reactant 1 and detection of a main peak with the desired m/z or the desired mass. Addition of NaHCO to the solution 3 Aqueous solution to adjust pH>7, concentrating the reaction mixture under reduced pressure to remove DCM and MeOH, filtering, and concentrating the filter cake under reduced pressure to give 3- [ 4-chloro-5- [4- (dimethoxymethyl) -1-piperidinyl as a white solid]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (232 mg,532.23umol,68.92% yield).
Step 3:
to 3- [ 4-chloro-5- [4- (dimethoxymethyl) -1-piperidinyl]-1-oxo-isoindolin-2-yl]To a solution of piperidine-2, 6-dione (232 mg,532.23umol,1 eq.) in THF (6 mL) was added HCl (2M, 3 mL). The mixture was stirred at 40℃for 0.5 h. LC-MS (EB 648-397-P1A) showed complete consumption of reactant 1 and detection To a main peak with the desired mass. Addition of NaHCO to the solution 3 Aqueous solution to adjust pH>7, followed by filtration and concentration of the filter cake under reduced pressure to give 1- [ 4-chloro-2- (2, 6-dioxo-3-piperidinyl) -1-oxo-isoindolin-5-yl as a yellow solid]Piperidine-4-carbaldehyde (118 mg,302.69umol,56.87% yield).
Step 4:
1- [ 4-chloro-2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl]Piperidine-4-carbaldehyde (50 mg,128.26umol,1 eq) and 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl]Pyrimidin-4-yl]A solution of 1H-indazole (57.41 mg,128.26umol,1 eq.) in AcOH (1 mL) and MeOH (10 mL) was stirred at 20deg.C for 20min, followed by the addition of borane; 2-methylpyridine (27.44 mg, 256.52. Mu. Mol,2 eq.). The mixture was stirred at 25℃under N 2 Stirring is carried out for 12 hours under an atmosphere. LC-MS (EB 648-398-P1A) showed complete consumption of reactant 1 and detection of a major peak with the desired mass. The solution was purified by preparative HPLC (Xtime C18X 30mm X10 um; water (FA) -CAN, B%:0% -35%,35 min) to give 3- [ 4-chloro-5- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl) as a white solid]Pyrimidin-4-yl ]-4-piperidinyl]Methyl group]Piperazin-1-yl]Methyl group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (59 mg,68.02umol,53.03% yield, 100% purity, FA).
Exemplary Synthesis of Compound 240
Step 1:
imidazole (1.33 g,19.58mmol,1 eq.) was added in one portion to a mixture of cyclopent-1, 3-diol (2 g,19.58mmol,1 eq.) and TBSCl (2.95 g,19.58mmol,2.40mL,1 eq.) in DCM (10 mL) at 0deg.C under N2. The mixture was stirred at 0 ℃ for 1 hour. TLC (petroleum ether: ethyl acetate=5/1, rf=0.35) showed that the reaction was complete. The reaction was quenched with saturated aqueous ammonium chloride (20 mL) followed by extraction with DCM (20 mL. Times.3). The combined organic layers were dried over Na2SO4, then filtered off and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=10/1) to give a mixture of 1,3-trans-3- [ tert-butyl (dimethyl) silyl ] oxycyclopentanol as a colorless liquid (1.14 g,5.27mmol,26.90% yield).
Step 2:
to 1,3-trans-3- [ tert-butyl (dimethyl) silyl at 0 ℃C]To a solution of oxycyclopentanol (1.5 g,6.93mmol,1 eq.) in THF (10 mL) were added TEA (1.05 g,10.40mmol,1.45mL,1.5 eq.) and TMSCl (828.39 mg,7.63mmol,967.74uL,1.1 eq.) and the reaction mixture was stirred at 25℃for 1 hour. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. To a stirred solution of the residue and benzyl 4-oxopiperidine-1-carboxylate (1.94 g,8.32mmol,1.66mL,1.2 eq.) in DCM (10 mL) at-60℃was then added Et dropwise 3 SiH (967.25 mg,8.32mmol,1.33mL,1.2 eq.) and TMSOTF (847.37 mg,3.81mmol,688.92uL,0.55 eq.) and the reaction mixture was stirred at 0deg.C under N 2 Stirred for 2 hours. LCMS showed the reaction was complete. The reaction mixture was quenched by addition of 30mL of water and extracted with DCM (20 mL x 3). The combined organic layers were washed with brine (30 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=5/1) to give 4- [ (1 s,3 s) -3- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy as a pale yellow liquid]Cyclopentyloxy group]Piperidine-1-carboxylic acid benzyl ester and 4- [ (1R, 3R) -3- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy]Cyclopentyloxy group]Benzyl piperidine-1-carboxylate (1.37 g,2.55mmol,36.83% yield).
/>
Step 3:
to a mixture of benzyl 4- [ (1 s,3 s) -3- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] cyclopentyloxy ] piperidine-1-carboxylate and benzyl 4- [ (1 r,3 r) -3- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] cyclopentyloxy ] piperidine-1-carboxylate (1.37 g,2.55mmol,1 eq.) in EtOH (10 mL) and EA (10 mL) at 25 ℃ was added Pd/C (500 mg,10% purity, 1.00 eq.) in one portion. The mixture was stirred at 25℃under H2 (15 psi) for 12 hours. TLC (petroleum ether: ethyl acetate=3/1) showed the reaction was complete. The mixture was filtered and concentrated under reduced pressure at 45 ℃. The residue was used in the next step without further purification. The product mixture was obtained as a pale yellow liquid 4- [ (1 s,3 s) -3- (4-piperidinyloxy) cyclopentyloxy ] piperidine and 4- [ (1 r,3 r) -3- (4-piperidinyloxy) cyclopentyloxy ] piperidine (880 mg, crude).
Step 4:
at N 2 Downward 4- [ (1S, 3S) -3- (4-piperidinyloxy) cyclopentyloxy]Piperidine and 4- [ (1R, 3R) -3- (4-piperidinyloxy) cyclopentyloxy]To a solution of piperidine (500 mg,1.86mmol,2 eq.) and 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (317.75 mg,931.47umol,1 eq.) in DMSO (4 mL) was added Cs 2 CO 3 (606.98 mg,1.86mmol,2 eq.) and PD-PEPSI (TM) -IPENT catalyst (73.93 mg,93.15umol,0.1 eq.). After addition, the mixture was stirred at 90℃under N 2 Stirred for 10 hours. LCMS showed the reaction was complete. The mixture was filtered and concentrated under reduced pressure. The residue was purified by prep HPLC (xtime C18 100 x 30mm x 10um; mobile phase: [ water (FA) -ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -35%,35 min) to give 3- [ 4-fluoro-1-oxo-5- [4- [ (1S, 3S) -3- (4-piperidinyloxy) cyclopentyloxy) as a grey solid]-1-piperidinyl group]Isoindolin-2-yl]Piperidine-2, 6-dione and 3- [ 4-fluoro-1-oxo-5- [4- [ (1R, 3R) -3- (4-piperidinyloxy) cyclopentyloxy]-1-piperidinyl group]Isoindolin-2-yl]Piperidine-2, 6-dione (130 mg,226.23umol,24.29% yield, FA).
Step 5:
at 25 ℃ at N 2 Downward 3- [ 4-fluoro-1-oxo-5- [4- [ (1S, 3S) -3- (4-piperidinyloxy) cyclopentyloxy) ]-1-piperidinyl group]Isoindolin-2-yl]Piperidine-2, 6-dione and 3- [ 4-fluoro-1-oxo-5- [4- [ (1R, 3R) -3- (4-piperidinyloxy) cyclopentyloxy]-1-piperidinyl group]Isoindolin-2-yl]DIEA (146.19 mg,1.13mmol,197.02uL,5 eq.) was added in one portion to a mixture of piperidine-2, 6-dione (130 mg,226.23umol,1 eq., FA) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -2H-indazole (68.04 mg,226.23umol,1 eq.) in DMSO (2 mL). The mixture was stirred at 80℃for 12 hours. LCMS showed the reaction was complete. The mixture was cooled to 25 ℃ and poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by prep HPLC (xtime C18 100 x 30mm x 10um; mobile phase: [ water (FA) -ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:15% -55%,35 min) to give 3- [ 4-fluoro-5- [4- [ (1 s,3 s) -3- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl) as a gray solid]Pyrimidin-4-yl]-4-piperidinyl]Oxy group]Cyclopentyloxy group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione and 3- [ 4-fluoro-5- [4- [ (1R, 3R) -3- [ [1- [6- [5- (1-methylcyclopropoxy) -2H-indazol-3-yl ] ]Pyrimidin-4-yl]-4-piperidinyl]Oxy group]Cyclopentyloxy group]-1-piperidinyl group]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (102.6 mg,128.11umol,56.63% yield, 99% purity).
Exemplary Synthesis of Compound 241
Step 1:
to a solution of 2-bromo-4-fluoro-benzaldehyde (1.00 g,4.93mmol,1.0 eq) and 4- (dimethoxymethyl) piperidine (0.78 g,4.93mmol,1.0 eq) in dimethyl sulfoxide (20.0 mL) was added diisopropylethylamine (1.91 g,14.78mmol,2.6mL,3.0 eq) and the mixture was stirred at 90 ℃ for 1 hour. LCMS showed the starting material was consumed and the desired mass was detected. The mixture was partitioned between ethyl acetate (100 mL) and water (100 mL). The separated organic layer was washed with water, dried over anhydrous sodium sulfate and evaporated to dryness. The crude product was purified by silica gel chromatography (10% -30% ethyl acetate/petroleum ether). 2-bromo-4- [4- (dimethoxymethyl) -1-piperidinyl ] benzaldehyde (1.50 g,4.38mmol,88% yield, 100% purity) was obtained as a yellow solid which was detected by HNMR (EW 29242-68-P1C 1).
Step 2:
to a solution of 3-aminopiperidine-2, 6-dione (0.81 g,4.91mmol,1.2 eq.) in methanol (20.0 mL) was added triethylamine (0.41 g,4.09mmol,0.6mL,1.0 eq.) and the mixture was stirred for 0.5 h. 2-bromo-4- [4- (dimethoxymethyl) -1-piperidinyl ] benzaldehyde (1.40 g,4.09mmol,1.0 eq.) and acetic acid (0.24 g,4.09mmol,0.23mL,1.0 eq.) were then added. After stirring at 25℃for 2 hours, 2-methylpyridine borane (0.87 g,8.18mmol,2.00 eq.) was added and the mixture stirred at 25℃for 1 hour. LCMS showed the starting material was consumed and the desired mass was detected. The mixture was purified by silica gel chromatography (1% -10% methanol/dichloromethane). The compound 3- [ [ 2-bromo-4- [4- (dimethoxymethyl) -1-piperidinyl ] phenyl ] methylamino ] piperidine-2, 6-dione (0.9 g,1.98mmol,48% yield, 100% purity) was obtained as an off-white solid, which was detected by HNMR (EW 29242-71-P1C 1).
Step 3:
to a solution of 3- [ [ 2-bromo-4- [4- (dimethoxymethyl) -1-piperidinyl ] phenyl ] methylamino ] piperidine-2, 6-dione (0.70 g,1.54mmol,1.0 eq.) in dimethylformamide (10.0 mL) were added [1, 1-bis (diphenylphosphino) ferrocene ] palladium (II) dichloride (0.34 g,0.46mmol,0.3 eq.) and diisopropylethylamine (0.59 g,4.62mmol,0.81mL,3.0 eq.) under nitrogen. The suspension was degassed and purged 3 times with carbon dioxide. LCMS showed some starting material was consumed and the desired mass was detected. The mixture was stirred at 80℃under carbon monoxide (50 Psi) for 12 hours. The mixture was purified by preparative HPLC (2% -35% acetonitrile +0.225% formic acid/water, 11 min). The compound 3- [6- [4- (dimethoxymethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (0.31 g,0.77mmol,50% yield, 100% purity) was obtained as a white solid, which was detected by HNMR (EW 29242-75-P1C 1).
Step 4:
to a solution of 3- [6- [4- (dimethoxymethyl) -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (0.25 g,0.62mmol,1.00 eq.) in tetrahydrofuran (2.5 mL) was added 2M sulfuric acid (0.06 g,0.62mmol,2.5mL,1.00 eq.) at 0 ℃. The mixture was stirred at 40℃for 1.5 hours. LCMS showed the starting material was consumed and the desired mass was detected. The mixture was diluted with water (2 mL), neutralized with solid sodium bicarbonate until no sodium bicarbonate evolved. The mixture was extracted twice with ethyl acetate (4 mL). The organic layer was washed with water, dried over anhydrous sodium sulfate and evaporated to dryness. The product 1- [2- (2, 6-dioxo-3-piperidinyl) -3-oxo-isoindolin-5-yl ] piperidine-4-carbaldehyde (0.22 g,0.62mmol,99% yield, 100% purity) was obtained as a white solid, which was detected by HNMR (EW 29242-90-P1C 1).
Step 5:
to a solution of 5- (1-methylcyclopropoxy) -3- [6- [4- (piperazin-1-ylmethyl) -1-piperidinyl ] pyrimidin-4-yl ] -2H-indazole (0.07 g,0.16mmol,1.0 eq) in methanol (2.0 mL) was added sodium acetate (0.06 g,0.78mmol,5.0 eq) and stirred for 0.5 hours, 1- [2- (2, 6-dioxo-3-piperidinyl) -3-oxo-isoindol-5-yl ] piperidine-4-carbaldehyde (0.06 g,0.17mmol,1.1 eq) and acetic acid (0.02 g,0.31mmol,0.1mL,2.0 eq) were added to the above mixture, and the mixture was stirred for 0.5 hours, borane was added; 2-methylpyridine (0.03 g,0.31mmol,2.00 eq.) and the mixture was stirred at 25℃for 1 hour. The mixture was purified by preparative HPLC (5% -29% acetonitrile +0.225% formic acid/water, 12 min). The product 3- [6- [4- [ [4- [ [1- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] -4-piperidinyl ] methyl ] piperazin-1-yl ] methyl ] -1-piperidinyl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (27 mg,0.03mmol,22% yield, 98% purity) was obtained as a pink solid.
Exemplary synthesis of compound 242: compound 242 was prepared in a similar manner to compounds 219 and 223.
Exemplary synthesis of compound 243: compound 243 was prepared in a similar manner to compounds 234 and 239.
Exemplary synthesis of compound 244: compound 244 was prepared in a similar manner to compounds 210 and 235.
Exemplary synthesis of compound 245: compound 245 was prepared in a similar manner to compounds 210 and 219.
Exemplary syntheses of compounds 246, 255, 256, 257, 262, 264, 265, 271, 278, 285, 287, and 288: compounds 246, 255, 256, 257, 262, 264, 265, 271, 278, 285, 287 and 288 were prepared in a similar manner to compound 211.
Exemplary syntheses of compounds 247 and 274: compounds 247 and 274 were prepared in a similar manner to compound 224.
Exemplary synthesis of compound 248: compound 248 was prepared in a similar manner to compounds 210 and 211.
Exemplary synthesis of compound 249: compound 249 was prepared in a similar manner to compounds 213 and 239.
Exemplary synthesis of compounds 250 and 251: compounds 250 and 251 were prepared in a similar manner to compounds 211 and 224.
Exemplary syntheses of compounds 252, 268, and 281: compounds 252, 268, and 281 were prepared in a similar manner to compounds 211 and 213.
Exemplary syntheses of compounds 253, 261, 282, and 283: compounds 253, 261, 282, and 283 were prepared in a similar manner to compounds 201 and 211.
Exemplary synthesis of compounds 254 and 259: compounds 254 and 259 were prepared in a similar manner to compound 213.
Exemplary syntheses of compounds 258 and 260: compounds 258 and 260 were prepared in a similar manner to compound 234.
Exemplary syntheses of compounds 263, 266, 267, 272, 276, and 284: compounds 263, 266, 267, 272, 276 and 284 were prepared in a similar manner to compound 181.
Exemplary synthesis of compound 269: compound 269 was prepared in a similar manner to compounds 213 and 219.
Exemplary syntheses of compounds 270 and 275: compounds 270 and 275 were prepared in a similar manner to compound 236.
Exemplary syntheses of compounds 273 and 280: compounds 273 and 280 were prepared in a similar manner to compounds 211 and 234.
Exemplary synthesis of compounds 277 and 279: compounds 277 and 279 were prepared in a similar manner to compounds 211 and 236.
Exemplary synthesis of compound 286: compound 286 was prepared in a similar manner to compounds 219 and 234.
Exemplary synthesis of compound 289: compound 289 is prepared in a similar manner to compound 181.
Exemplary synthesis of compound 290: compound 290 was prepared in a similar manner to compound 181 using intermediate 4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperidine-1-carboxylic acid tert-butyl ester.
Step 1:
to a solution of 4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (2.72 g,8.79mmol,1 eq.) and 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (3 g,8.79mmol,1 eq.) in dioxane (30 mL) and H2O (3 mL) were added CsF (4.01 g,26.38mmol,972.72ul,3 eq.) and di-tert-butyl (cyclopentyl) phosphorane; palladium dichloride; iron (573.16 mg,879.42umol,0.1 eq). The mixture was stirred at 90 ℃ under nitrogen atmosphere for 16 hours. LCMS showed the desired MS. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel40g/>Silica gel flash column, eluent: 0% -100% ethyl acetate/petroleum ether gradient, 60 mL/min). Obtaining the compound 4- [2- (2, 6-dioxo-3-piperidyl) -4-fluoro-1-oxo-isoindolin-5-yl) as a white solid]-3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (3.7 g,8.34mmol,94.87% yield).
Step 2:
to 4- [2- (2, 6-dioxo-3-piperidyl) -4-fluoro-1-oxo-isoindolin-5-yl under N2 atmosphere]To a solution of tert-butyl 3, 6-dihydro-2H-pyridine-1-carboxylate (1.7 g,3.83mmol,1 eq.) in DMF (20 mL) was added Pd/C (2 g,3.83mmol,225.50uL,10% purity, 1.00 eq.). The suspension was degassed and used with H 2 Purging three times. The mixture was stirred at 40℃under H 2 (50 Psi) for 16 hours. LCMS showed the desired MS. Filtering the reaction mixture and concentrating under reduced pressure to obtainThe residue was taken up without purification. Obtaining the compound 4- [2- (2, 6-dioxo-3-piperidyl) -4-fluoro-1-oxo-isoindolin-5-yl) as an off-white solid]Tert-butyl piperidine-1-carboxylate (1.23 g,2.76mmol,72.03% yield).
Exemplary synthesis of compound 291: compound 291 was prepared in an analogous manner to compound 209 using intermediate 4- [ [ (2 s,5 r) -5- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] -1, 4-dioxane-2-yl ] methyl ] piperazine-1-carboxylic acid tert-butyl ester.
Step 1:
HgO (50.04 g,231.04mmol,1 eq.) was dissolved in HNO 3 (45.85 g,728.21mmol,32.75mL,3.15 eq.) and H 2 O (20 mL). Adding additional H 2 O (80 mL) and the solution was cooled to 0deg.C. Then, with stirring and cooling, prop-2-en-1-ol (28.03 g,482.62mmol,32.82mL,2.09 eq.) was added. A large amount of white precipitate gradually formed. The mixture was allowed to stand for 6 hours. (in this case, the reaction mixture may be left to stand for a long period of time to cause decomposition of the product). The precipitate was filtered off and dissolved in 10% aqueous sodium hydroxide. Concentrated aqueous potassium iodide was then added until trans-2, 5-bis- (iodomercuric methyl) -p-dioxane was completely precipitated. It is filtered off and washed with water. Combining wet solids with I 2 (117.28 g,462.07mmol,93.08mL,2 eq.) and KI (106.53 g,642.28mmol,2.78 eq.) in H 2 A solution in O (500 mL) water was refluxed. (it is recommended to use a flask large enough that the reactants make up only about one third of their volume, toluene (20 mL) is added to wash back the sublimated iodine on the condenser.) reflux (110 ℃) is continued until the organomercury compound has reacted completely (about 48 hours). TLC (petroleum ether: ethyl acetate=10:1) showed a new spot. After cooling, the reaction mixture was extracted with ethyl acetate (400 ml x 3). The organic layer was washed with sodium bisulphite solution (300 ml x 3), dried over sodium sulphate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether),trans-2, 5-bis (iodomethyl) -1, 4-dioxane (24 g,65.23mmol,28.23% yield) was obtained as a white solid.
Step 2:
to a solution of trans-2, 5-bis (iodomethyl) -1, 4-dioxane (1.7 g,4.62mmol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester (774.46 mg,4.16mmol,0.9 eq.) in MeCN (6 mL) was added TEA (1.87 g,18.48mmol,2.57mL,4 eq.) and the reaction was taken at 80℃under N 2 Stirred for 16 hours. TLC (petroleum ether: ethyl acetate=0:1, pma) of the reaction showed a new spot, and LCMS showed detection of the desired MS. Passing the reaction through H 2 The O (40 mL) solution was quenched and extracted with ethyl acetate (3 x 40 mL). The combined organic phases were washed with water, with Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=1:1) to give 4- [ [ (2 r,5 s) -5- (iodomethyl) -1, 4-dioxan-2-yl as a brown solid]Methyl group]Piperazine-1-carboxylic acid tert-butyl ester (210 mg,492.62 mol,10.66% yield).
Step 3:
to 4- [ [ (2 r,5 s) -5- (iodomethyl) -1, 4-dioxan-2-yl at 25℃under N2]Methyl group]To a mixture of tert-butyl piperazine-1-carboxylate (340 mg,797.58umol,1 eq) and 3- (4-fluoro-1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (276.25 mg,797.58umol,1 eq) in DMSO (2 mL) was added DIEA (206.16 mg,1.60mmol,277.85uL,2 eq) in one portion. The mixture was heated to 100 ℃ and stirred for 12 hours. TLC (petroleum ether: ethyl acetate=0:1) and LCMS showed the reaction was complete. Passing the reaction through H 2 The O (40 mL) solution was quenched and extracted with ethyl acetate (3 x 40 mL). The combined organic phases were washed with water, with Na 2 SO 4 Drying and concentrating in vacuum to obtainResidue. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=0/1) to give 4- [ [ (2 s,5 r) -5- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a dark brown solid ]Piperazin-1-yl]Methyl group]-1, 4-dioxan-2-yl]Methyl group]Tert-butyl piperazine-1-carboxylate (180 mg,279.19umol,35.00% yield). The compounds are prepared as mixtures of trans dioxane diastereomers.
Exemplary synthesis of compound 292: compound 292 was prepared in a similar manner to compounds 181 and 290 using intermediate 4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester.
Exemplary synthesis of compound 293: compound 293 was prepared in a similar manner to compound 211 using the intermediate trans- [3- (4-piperidinyloxy) cyclobutyl ] methanol.
Exemplary Synthesis of the intermediate trans- [3- (4-piperidinyloxy) cyclobutyl ] methanol
Step 1:
to a solution of methyl 3-hydroxycyclobutane formate (3.7 g,28.43mmol,1 eq.) in THF (10 mL) at 0deg.C were added TEA (3.31 g,32.70mmol,4.55mL,1.15 eq.) and TMSCL (3.40 g,31.27mmol,3.97mL,1.1 eq.) and the reaction mixture was stirred at 20deg.C for 30 min. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue. To a stirred solution of the residue and benzyl 4-oxopiperidine-1-carboxylate (7.29 g,31.27mmol,6.24mL,1.1 eq.) in DCM (10 mL) was then added Et3SiH (3.80 g,32.70mmol,5.22mL,1.15 eq.) and TMSOTF (3.16 g,14.22mmol,2.57mL,0.5 eq.) dropwise at-60℃and the reaction mixture was stirred at 0℃under N 2 Stirred for 1.5 hours. LCMS showed the desired MS. TLC (petroleum ether: ethyl acetate=3:1) showed several new spots. The reaction mixture was purified by adding saturated NaHCO at 0deg.C 3 (ph=8) followed by extraction dilution with DCM (20 ml×3). Will be combinedThe combined organic layers were washed with brine (15 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel (0% -16% ethyl acetate/petroleum ether) to give benzyl 4- (3-methoxycarbonylcyclobutoxy) piperidine-1-carboxylate (5 g,11.08mmol,38.98% yield, 77% purity) as a colorless oil.
Step 2:
at 25 ℃ at H 2 To a mixture of benzyl 4- (3-methoxycarbonylcyclobutoxy) piperidine-1-carboxylate (5 g,14.39mmol,1 eq.) in EtOH (10 mL) was added Pd/C (3 g,14.39mmol,10% purity, 1 eq.) over 2 hours (15 PSI). TLC (petroleum ether: ethyl acetate=3:1) showed a new major spot. The resulting product was concentrated in vacuo to give methyl 3- (4-piperidinyloxy) cyclobutanecarboxylate (3.1 g, crude) as a colorless oil.
Step 3:
at 0 ℃ at N 2 To a solution of methyl 3- (4-piperidinyloxy) cyclobutanecarboxylate (3 g,14.07mmol,1 eq.) in THF (10 mL) was added LiAlH4 (1.07 g,28.13mmol,2 eq.) in portions. After the addition, the reaction mixture was stirred at 0 ℃ for 1 hour. TLC (dichloromethane: methanol=5:1, rf=0.07) showed no starting material and a new spot. By adding H 2 O (1 mL), followed by 15% aqueous NaOH (1 mL) and water (3 mL) quench the reaction mixture. After stirring at room temperature for 30min, the mixture was filtered through a pad of celite to remove solids. The filtrate was concentrated to dryness to give ((1 r,3 r) -3- (piperidin-4-yloxy) cyclobutyl) methanol (2 g,10.80mmol,76.75% yield) as a white solid.
Exemplary synthesis of compound 294: compound 294 was prepared in an analogous manner to compound 181 using intermediate (3 s,4 r) -3-fluoro-4-formyl-piperidine-1-carboxylic acid benzyl ester.
Step 1:
to a solution of (3-fluoro-4-pyridinyl) methanol (14.8 g,116.43mmol,1 eq.) in acetone (130 mL) was added BnBr (21.90 g,128.07mmol,15.21mL,1.1 eq.). The mixture was stirred at 65℃for 16 hours. TLC (petroleum ether: ethyl acetate=0:1, rf=0.1) showed new spots. The reaction mixture was cooled to room temperature and diluted with MTBE (100 ml). The suspension was filtered, followed by stirring and filtration of the wet cake with 25% acetone/MTBE v/v (200 ml). The filtrate was then dried in vacuo to give (1-benzyl-3-fluoro-pyridin-1-ium-4-yl) methanol (25 g,114.55mmol,98.39% yield) as a yellow solid.
Step 2:
to a solution of (1-benzyl-3-fluoro-pyridin-1-ium-4-yl) methanol (25 g,114.55mmol,1 eq.) in MeOH (300 mL) at 0deg.C was then added NaBH at 0deg.C 4 (6.50 g,171.82mmol,1.5 eq.). The mixture was stirred at 0 ℃ for 2 hours. TLC (petroleum ether: ethyl acetate=0:1, rf=0.6) showed new spots. LCMS showed the desired MS. The reaction mixture was treated with 100mL NH 4 C1 (saturation) quenching. The aqueous phase was extracted with ethyl acetate (3X 100 mL). The combined organic layers were washed with brine (3×80 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel330g/>Silica gel flash column, eluent: 0% -100% ethyl acetate/petroleumEther gradient, 80 mL/min) to give (1-benzyl-5-fluoro-3, 6-dihydro-2H-pyridin-4-yl) methanol (13 g,58.75mmol,51.29% yield) as a yellow gum.
Step 3:
at N 2 Cyclopentane was charged to a three-necked round bottom flask under atmosphere; dicyclohexyl- [ (1R) -1- [2- (2-diphenylphosphoranylphenyl) cyclopentyl]Ethyl group]A phosphorane; iron (22.5 mg,33.55umol,1.48e-3 eq.) chlororhodium; (1Z, 5Z) -cycloocta-1, 5-diene (7.5 mg,15.21umol,6.73e-4 eq.) and dry nitrogen degassed DCM (10 mL). The solution was stirred at ambient temperature for 45 minutes. A solution of (1-benzyl-5-fluoro-3, 6-dihydro-2H-pyridin-4-yl) methanol (5 g,22.60mmol,1 eq.) in anhydrous MeOH (50 mL) was added to a nitrogen purged 250mL stainless steel pressure vessel. Subsequently, the above aged catalyst solution was added to the vessel under a nitrogen stream. Subjecting the resulting mixture to H 2 Degassing three times, followed by stirring at 50℃under 2.0MPa H 2 The mixture was heated under (2.0 MPa, about 300 psi) for 36 hours. LCMS showed all desired products. TLC (petroleum ether: ethyl acetate=0:1, rf=0.3) showed new spots. The reaction vessel was cooled to room temperature and purged with nitrogen. The reaction mixture was concentrated in vacuo to give a dark brown oil. The concentrate was dissolved in EtOAc (50 mL) and saturated NaHCO was added 3 Aqueous solution (50 mL). The mixture was stirred at room temperature for half an hour and the organic phase was separated. The aqueous phase was extracted three times with EtOAc (60 ml). The combined organic phases were washed with brine, dried over Na 2 SO 4 Dried and concentrated under vacuum. The residue was purified by flash chromatography on silica gel330g/>Silica gel flash column, eluent: gradient of 0% -100% ethyl acetate/petroleum ether, 60 mL/min) to give a yellow gum((3 r,4 s) - (1-benzyl-3-fluoro-4-piperidinyl) methanol (2.95 g,13.21mmol,58.47% yield))>
Step 4:
to a stirred solution of ((3 r,4 s) -1-benzyl-3-fluoropiperidin-4-yl) methanol (2.95 g,13.21mmol,1 eq.) in MeOH (10 mL) was added Pd/C (300 mg,13.21mmol,10% purity, 1 eq.) and HCl (6M, 3mL,1.36 eq.). The mixture was purged three times with hydrogen at 15 psi. After stirring at 50 ℃ for 6 hours, the starting material was consumed. LCMS showed no starting material. TLC (petroleum ether: ethyl acetate=0:1, rf=0.01) showed new spots. The reaction mixture was then filtered through celite, the pad was rinsed with MeOH, and the filtrate was concentrated in vacuo to give [ (3 s,4 r) -3-fluoro-4-piperidinyl ] methanol (2.2 g, crude, HCl) as a white solid.
Step 5:
to [ (3 s,4 r) -3-fluoro-4-piperidinyl group at 0 ℃C]To a solution of methanol (2.2 g,12.97mmol,1.18 eq, HCl) in DCM (50 mL) was added TEA (5.56 g,54.96mmol,7.65mL,5 eq) and stirred at 0deg.C for 30 min. CbzCl (3.60 g,21.10mmol,3mL,1.92 eq.) was then added and the mixture was taken at 0deg.C under N 2 Stirred for 2h. LCMS showed the desired product. TLC of the reaction (petroleum ether: ethyl acetate=0:1, rf=0.5) showed a new spot. The reaction was diluted with water (40 mL). The mixture was extracted with ethyl acetate (50 ml x 3), dried over Na 2 SO 4 Dried and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (0% -100% ethyl acetate/petroleum ether) to give benzyl (3 s,4 r) -3-fluoro-4- (hydroxymethyl) piperidine-1-carboxylate (2.8 g,10.48mmol,95.30% yield) as a colorless gum.
Step 6:
to a solution of benzyl (3 s,4 r) -3-fluoro-4- (hydroxymethyl) piperidine-1-carboxylate (500 mg,1.87mmol,1 eq.) in DCM (10 mL) at 20deg.C was added DMP (1000.00 mg,2.36mmol,729.93uL,1.26 eq.) and stirred at 20deg.C for 1 hour. LCMS showed the desired product. TLC of the reaction (petroleum ether: ethyl acetate=0:1, rf=0.6) showed a new spot. The reaction mixture was purified by adding saturated NaHCO at 0deg.C 3 (pH adjusted about 8) and extracted with DCM (20 ml x 3). The combined organic layers were treated with saturated NaSO 3 (20 ml x 2) and brine (10 ml x 2), washed with Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give (3 s,4 r) -3-fluoro-4-formyl-piperidine-1-carboxylic acid benzyl ester (450 mg, crude) as a colourless gum.
Exemplary synthesis of compound 295: compound 295 was prepared in a similar manner to compound 181 starting from tert-butyl (3S) -3- (hydroxymethyl) pyrrolidine-1-carboxylate.
Exemplary synthesis of compound 296: compound 296 was prepared in a similar manner to compound 181 using the intermediate tert-butyl 4- [ [ (2 r,5 s) -5- (iodomethyl) -1, 4-dioxan-2-yl ] methyl ] piperazine-1-carboxylate.
Exemplary synthesis of compound 297: compound 297 was prepared in an analogous manner to compound 181 using intermediate (3 r,4 s) -4- (2, 6-dioxopiperidin-3-yl) -4-fluoro-1-oxoisoindolin-5-yl) -3-fluoropiperidine-1-carboxylic acid tert-butyl ester.
Step 1:
to 4- [2- (2, 6-dioxo-3-piperidyl) -4-fluoro-1-oxo-isoindolin-5-yl at 0 ℃C]To a solution of tert-butyl-3, 6-dihydro-2H-pyridine-1-carboxylate (4 g,9.02mmol,1 eq.) in THF (400 mL) was added drop wise BH 3 -Me 2 S (10M, 9.02mL,10 eq). After the addition, the mixture was stirred at 25℃for 1 2 hours, followed by dropwise addition of sodium 3-dioxaborane oxide tetrahydrate (4.16 g,27.06mmol,5.20mL,3 eq.) in H at 0deg.C 2 O (20 mL). The resulting mixture was stirred at 25℃for 10 hours. LCMS showed the detection of the desired mass. The reaction mixture was quenched with saturated sodium sulfite solution (80 mL), and the mixture was extracted with ethyl acetate (70 ml×3). The combined organic phases were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography on silica gel40g/>Silica gel flash column, eluent: purification with a 0% -15% methanol/dichloromethane gradient, 80mL/min, afforded 4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl) as a white solid]-3-hydroxy-piperidine-1-carboxylic acid tert-butyl ester (1.5 g,2.67mmol,29.55% yield, 82% purity).
Step 2:
to a solution of tert-butyl 4- (2, 6-dioxopiperidin-3-yl) -4-fluoro-1-oxoisoindolin-5-yl) -3-hydroxypiperidine-1-carboxylate (1.5 g,3.25mmol,1 eq.) in DCM (100 mL) was added N-ethyl-N- (trifluoro- λ4-sulfanyl) ethylamine (2.10 g,13.00mmol,1.72mL,4 eq.) at-78deg.C. The mixture was stirred at 20℃for 1 hour. TLC (dichloromethane: methanol=10:1) indicated complete consumption of reactant 1 and formation of new spots. The reaction mixture is reacted with H 2 O (20 mL) was mixed and extracted with DCM (20 mL. Times.3). The combined organic layers were washed with brine, dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel40g/>Silica gel flash column, eluent: 0% -79% ethyl acetate/petroleum ether gradient, 100 mL/min) to give (3 r,4 s) -4- (2, 6-dioxopiperidin-3-yl) -4-fluoro-1-oxoisoindolin-5-yl) -3-fluoropiperidine-1-carboxylic acid tert-butyl ester (627 mg,1.35mmol,41.62% yield) as a white solid.
Exemplary synthesis of compound 298: compound 298 was prepared in a similar manner to compound 211 using the intermediate tert-butyl 3- (4-piperidinyloxy) azetidine-1-carboxylate.
Exemplary synthesis of compound 299: compound 299 was prepared in a similar manner to compound 181 using the intermediate benzyl (3 s,4 r) -3-fluoro-4- (hydroxymethyl) piperidine-1-carboxylate.
Exemplary synthesis of compound 300: compound 300 was prepared in a similar manner to compound 209 using the intermediate trans- [3- (4-piperidinyloxy) cyclobutyl ] methanol.
Exemplary synthesis of compound 301: compound 301 was prepared in a similar manner to compound 209 using the intermediate tert-butyl 3-fluoro-3- (hydroxymethyl) azetidine-1-carboxylate.
Exemplary synthesis of compound 302: compound 302 was prepared in a similar manner to compound 211 using intermediate 4- [ [ (2 r,5 s) -5- (iodomethyl) -1, 4-dioxan-2-yl ] methyl ] piperazine-1-carboxylic acid tert-butyl ester and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine.
Exemplary synthesis of compound 303: compound 303 was prepared in a similar manner to compound 181 starting from tert-butyl (3S) -3- (hydroxymethyl) pyrrolidine-1-carboxylate.
Exemplary synthesis of compound 304: compound 304 was prepared in a similar manner to compound 181 using intermediate 4- ((1 r,3 r) -3- (2, 7-diazaspiro [3.5] non-7-yl) cyclobutoxy) piperidine-1-carboxylic acid tert-butyl ester.
Step 1:
to 2, 7-diazaspiro [3.5] at 0deg.C]To a solution of tert-butyl nonan7-carboxylate (3 g,13.26mmol,1 eq.) in DCM (30 mL) was added CbzCl (2.88 g,16.88mmol,2.4mL,1.27 eq.) and TEA (4.02 g,39.77mmol,5.54mL,3 eq.). The mixture was stirred at 20℃for 2 hours. LC-MS shows that the reactants are consumed and there is a new main peak with the desired m/z. The reaction mixture was treated with H 2 O (50 mL) was diluted and extracted with DCM (20 mL. Times.3). The combined organic layers were washed with brine, dried over Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel40g/>Silica gel flash column, eluent: gradient of 0% -32% ethyl acetate/petroleum ether, 100 mL/min) to give 2, 7-diazaspiro [3.5] as a colorless oil]Nonane-2, 7-dicarboxylic acid O2-benzyl ester O7-tert-butyl ester (2.77 g,7.68mmol,57.97% yield).
Step 2:
to a solution of 2-benzyl 2, 7-nonane-2, 7-dicarboxylic acid O7-tert-butyl ester (2.77 g,7.68mmol,1 eq.) in DCM (10 mL) was added TFA (7.70 g,67.53mmol,5.00mL,8.79 eq.). The mixture was stirred at 20℃for 30 minutes. LC-MS shows that the reactants are consumed and there is a new main peak with the desired m/z. The reaction mixture was concentrated under reduced pressure to remove DCM to give benzyl 2, 7-diazaspiro [3.5] nonane-2-carboxylate (3.2 g, crude, TFA) as a yellow oil.
Step 3:
to 2, 7-diazaspiro [3.5]]Nonane-2-carboxylic acid benzyl ester (1.25 g,4.80mmol,1 eq.) and 4- [3- (trifluoromethylsulfonyloxy) cyclobutoxy]To a solution of tert-butyl piperidine-1-carboxylate (2.52 g,6.24mmol,1.3 eq.) in MeCN (10 mL) was added DIEA (3.10 g,24.01mmol,4.18mL,5 eq.). The mixture was stirred at 60℃for 12 hours. LCMS showed the reaction was consumed and a new main peak with the desired m/z was detected. Silica powder was added to the reaction mixture and concentrated under reduced pressure to remove MeCN. The residue was purified by flash chromatography on silica gel 40g/>Silica gel flash column, eluent: 0% -8% methanol in dichloromethane gradient, 100 mL/min) to afford 7- [3- [ (1-tert-butoxycarbonyl-4-piperidinyl) oxy ] as a yellow solid]Cyclobutyl group]-2, 7-diazaspiro [3.5]]Nonane-2-carboxylic acid benzyl ester (2 g,3.89mmol,81.09% yield).
Step 4:
a mixture of 7- [3- [ (1-tert-butoxycarbonyl-4-piperidinyl) oxy ] cyclobutyl ] -2, 7-diazaspiro [3.5] nonane-2-carboxylic acid benzyl ester (1 g,1.95mmol,1 eq) and Pd/C (200 mg,1.95mmol,10% purity, 1 eq) in EtOH (10 mL) was degassed and purged 3 times with H2, then the mixture was stirred under H2 atmosphere at 20℃for 2 hours. TLC (dichloromethane: methanol=10:1) indicated that the reaction was consumed and a new spot formed. According to TLC, the reaction was clean. To the reaction mixture was added diatom ooze, filtered and concentrated under reduced pressure to give tert-butyl 4- ((1 r,3 r) -3- (2, 7-diazaspiro [3.5] non-7-yl) cyclobutoxy) piperidine-1-carboxylate (526 mg, crude) as a colorless oil.
Exemplary synthesis of compound 305: compound 305 was prepared in a similar manner to compound 181 using the intermediate tert-butyl 4- [ [ (3 r,6 r) -6- (piperazin-1-ylmethyl) tetrahydropyran-3-yl ] methyl ] piperazine-1-carboxylate.
Step 1:
to a stirred solution of diethyl malonate (100 g,624.35mmol,94.34mL,1 eq.) in THF (800 mL) at 0 ℃ was added NaH (27.47 g,686.78mmol,60% purity, 1.1 eq.) in portions. The mixture was stirred at 0 ℃ for 30 minutes. 4-Bromobut-1-ene (92.72 g,686.78mmol,69.71mL,1.1 eq.) was added to the mixture. The resulting mixture was stirred at 50℃for 12 hours. TLC (petroleum ether: ethyl acetate=4:1) showed the formation of new spots as the main product. Pouring the mixture into cold saturated NH 4 Aqueous Cl (1L) and the aqueous phase was extracted with ethyl acetate (600 ml x 3). The combined organic layers were dried over anhydrous Na 2 SO 4 Dried, filtered, and the filtrate concentrated in vacuo. Passing the residue through MPLCX g/>Silica gel flash column, eluent: 0% -10% ethyl acetate/petroleum ether gradient, 90 mL/min). Diethyl 2-but-3-enylmalonate (72 g,336.04mmol,53.82% yield) was obtained as a colorless oil.
Step 2:
to a stirred solution of diethyl 2-but-3-enylmalonate (72 g,336.04mmol,1 eq.) in t-BuOH (400 mL) and MeOH (80 mL) at 25℃was added NaBH in portions 4 (38.71 g,1.02mol,3.05 eq.). The resulting mixture was stirred at 25℃for 12 hours. TLC (PE: ea=2:1) showed reaction The material is consumed and a new spot is formed. Hydrochloric acid (1N) was added until the solution was neutral at 0 ℃. The aqueous phase was extracted with ethyl acetate (300 ml x 3). The combined organic layers were dried over anhydrous Na 2 SO 4 Dried, filtered, and the filtrate concentrated in vacuo. The residue was used in the next step without further purification. 2-but-3-enylpropane-1, 3-diol (42 g,322.62mmol,96.01% yield) was obtained as a pale yellow oil.
Step 3:
iodine (122.83 g,483.93mmol,97.48mL,1.5 eq.) and NaHCO at 0deg.C 3 To a stirred suspension of (40.65 g,483.93mmol,18.82mL,1.5 eq.) in tetrahydrofuran (200 mL) and water (80 mL) was added a solution of 2-but-3-enylpropane-1, 3-diol (42 g,322.62mmol,1 eq.) in THF (200 mL). The mixture was stirred at 25℃for 12 hours. LCMS showed the detection of the desired mass. Saturated Na at 0 DEG C 2 S 2 O 3 Aqueous solution (500 mL) was slowly added to the mixture, and the mixture was stirred at 25 ℃ for 10 minutes. The solution was extracted with EtOAc (300 ml x 3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (PE: etoac=100:1-1:1). [6- (iodomethyl) tetrahydropyran-3-yl ] as a pale yellow oil ]Methanol (34 g,132.77mmol,41.15% yield).
Step 4:
to [6- (iodomethyl) tetrahydropyran-3-yl]To a stirred solution of methanol (12.4 g,48.42mmol,1 eq.) in DMF (100 mL) was added K 2 CO 3 (16.73 g,121.06mmol,2.5 eq.) and benzyl piperazine-1-carboxylate (12.80 g,58.11mmol,11.23mL,1.2 eq.). The reaction mixture was stirred at 100℃for 12 hours. LCMS showed detection of the desiredQuality. Water (300 mL) was added to the mixture and the aqueous phase was extracted with ethyl acetate (200 mL x 3). The combined organic layers were washed with brine (200 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered, and the filtrate concentrated in vacuo. The residue was purified by silica gel chromatography (DCM: meoh=100:1-10:1). 4- [ [5- (hydroxymethyl) tetrahydropyran-2-yl ] as a mixture of diastereomers is obtained as a colorless oil]Methyl group]Benzyl piperazine-1-carboxylate (13 g,37.31mmol,77.05% yield).
Step 5:
it was determined by SFC that cis isomer 4- (((2 r,5 s) -5- (hydroxymethyl) tetrahydro-2H-pyran-2-yl) methyl) piperazine-1-carboxylic acid benzyl ester or its enantiomer (1.53 g,3.92mmol,11.01% yield, 89.24% purity, peak 4:de=100%, rt=1.416 min) was obtained by SCF purification of a diastereomer mixture of 4- [ [5- (hydroxymethyl) tetrahydro-2-yl ] methyl ] piperazine-1-carboxylic acid benzyl ester (12.40 g,35.59mmol,1 eq). The first fraction (10.19 g,29.25mmol,82.19% yield) obtained was a mixture of the single cis isomer (peak 2: rt= 1.161 min) and the trans isomer (peak 1: rt=1.125 min, peak 3: rt=1.195 min).
Step 6:
by SFC resolution of a diastereomer mixture of 1 cis isomer and 2 trans isomers (10.19 g,29.25 mmol), benzyl 4- [ [ (2 r,5 r) -5- (hydroxymethyl) tetrahydropyran-2-yl ] methyl ] piperazine-1-carboxylate or its enantiomer (trans isomer confirmed by HSQC, peak 3: de% = 100, rt = 2.614min,3.24g,8.86mmol,30.31% yield, 95.32% purity) was obtained as a pale yellow oil. The remaining diastereomer mixture (mixture of cis-isomer 2 and trans-isomer 2, 4.19g,12.03mmol,41.14% yield, peak 1:rt=1.156 min, peak 2:rt=1.244 min) was obtained as a pale yellow oil.
Step 7:
the cis product 4- [ [ (2 s,5 s) -5- (hydroxymethyl) tetrahydropyran-2-yl ] methyl ] piperazine-1-carboxylic acid ester or enantiomer thereof (2.41 g,6.58mmol,54.71% yield, 95.11% purity, yellow oil, cis isomer 2, peak 1:de=100%, rt= 1.977 min) and trans product 4- [ [ (2 s,5 r) -5- (hydroxymethyl) tetrahydropyran-2-yl ] methyl ] piperazine-1-carboxylic acid benzyl ester or enantiomer thereof (3.10 g,8.45mmol,70.27% yield, 94.98% purity, yellow oil, trans isomer 2, peak 2:de=100%, rt= 2.088 min) were obtained by SFC separation of the trans and cis diastereomers.
/>
Step 8:
to a solution of benzyl 4- [ [ (2 s,5 s) -5- (hydroxymethyl) tetrahydropyran-2-yl ] methyl ] piperazine-1-carboxylate or its enantiomer (780 mg,2.55mmol,1 eq.) in DCM (2 mL) was added Dess-Martin periodinane (1.08 g,2.55mmol,790.78ul,1 eq.). The mixture was stirred at 20℃for 12 hours. TLC plates showed no starting material remained and a new spot of less polarity formed. The reaction mixture was diluted with water (10 mL), neutralized to ph=7 with sodium bicarbonate, followed by extraction with 15mL (5 mL x 3) of DCM. The combined organic phases were dried over anhydrous sodium sulfate and concentrated in vacuo to give a residue without further purification. The compound 4- [ [ (2 s,5 r) -5-formyltetrahydropyran-2-yl ] methyl ] piperazine-1-carboxylic acid benzyl ester or its enantiomer (750 mg,2.17mmol,84.76% yield) was obtained as a yellow solid.
Step 9:
to a solution of tert-butyl piperazine-1-carboxylate (578.61 mg,2.60mmol,1.2 eq, HCl) in DMF (5 mL) was added NMM (437.97 mg,4.33mmol,476.05uL,2 eq) followed by benzyl 4- [ [ (2S, 5R) -5-formyltetrahydropyran-2-yl ] methyl ] piperazine-1-carboxylate or its enantiomer (750 mg,2.17mmol,1 eq) and sodium borohydride acetate (917.71 mg,4.33mmol,2 eq). The mixture was stirred at 20℃for 3 hours. TLC indicated that a major new spot of greater polarity was detected. The reaction mixture was partitioned between 10mL of water and 30mL of DCM (10 mL x 3). The organic phase was separated, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by preparative TLC (SiO 2, etOAc: pe=1:2) to give tert-butyl 4- [ [ (3 s,6 s) -6- [ (4-benzyloxycarbonylpiperazin-1-yl) methyl ] tetrahydropyran-3-yl ] methyl ] piperazine-1-carboxylate, or an enantiomer thereof, as a yellow solid (900 mg,1.74mmol,80.46% yield).
Step 10:
4- [ [ (3S, 6S) -6- [ (4-Benzyloxycarbonylpiperazin-1-yl) methyl]Tetrahydropyran-3-yl]Methyl group]A mixture of tert-butyl piperazine-1-carboxylate or its enantiomer (900 mg,1.74mmol,1 eq.) and Pd/C (90 mg,10% purity) in i-PrOH (10 mL) was degassed and H 2 Three purges followed by 25 ℃ of the mixture at H 2 Stirring is carried out for 2 hours under an atmosphere. LC-MS showed no residue of the reaction. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The compound 4- [ [ (3 s,6 s) -6- (piperazin-1-ylmethyl) tetrahydropyran-3-yl was obtained as a white solid without further purification]Methyl group]Piperazine-1-carboxylic acid tert-butyl ester or its enantiomer (560 mg,1.46mmol,84.04% yield).
Step 11:
to 4- [ [ (3S, 6S) -6- (piperazin-1-ylmethyl) tetrahydropyran-3-yl]Methyl group]To a solution of tert-butyl piperazine-1-carboxylate or its enantiomer (500 mg,1.31mmol,1.1 eq) and 3- (5-bromo-4-fluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (405.34 mg,1.19mmol,1 eq) in DMF (2 mL) was added cesium carbonate (619.44 mg,1.90mmol,1.6 eq). After addition, the reaction was taken up in N 2 Washing 3 times, followed by addition of (SP-4-1) - [1, 3-bis [2, 6-bis (1-ethylpropyl) phenyl ] ]-4, 5-dichloro-1, 3-dihydro-2H-imidazol-2-ylidene]Dichloro (3-chloropyridine-. Kappa.N) Palladium (102.25 mg,0.12mmol,0.1 eq.) the mixture was stirred at 90℃under N 2 Stirring is carried out for 16 hours under an atmosphere. LC-MS showed detection of the desired compound. The reaction mixture was filtered, then the filtrate was adjusted to ph=7 with formic acid and concentrated under vacuum to give a residue. The residue was purified by preparative HPLC (column: phenomenex Synergi Polar-RP 100X 25mm X4 um; mobile phase: [ water (TFA) -ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B%:17% -37%,7 min) to give 4- [ [ (3 s,6 s) -6- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] as a yellow solid]Piperazin-1-yl]Methyl group]Tetrahydropyran-3-yl]Methyl group]Piperazine-1-carboxylic acid tert-butyl ester or its enantiomer (150 mg,233.37 mol,19.64% yield).
Step 12:
to a solution of tert-butyl 4- [ [ (3 s,6 s) -6- [ [4- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] piperazin-1-yl ] methyl ] tetrahydropyran-3-yl ] methyl ] piperazine-1-carboxylate or enantiomer thereof (60 mg,0.093mmol,1 eq.) in DCM (2 mL) was added TFA (770.00 mg,6.75mmol,0.5mL,72.34 eq.). The mixture was stirred at 25℃for 15 minutes. TLC showed that new spots formed. The reaction mixture was concentrated under reduced pressure to give 3- [ 4-fluoro-1-oxo-5- [4- [ [ (2 s,5 s) -5- (piperazin-1-ylmethyl) tetrahydropyran-2-yl ] methyl ] piperazin-1-yl ] isoindolin-2-yl ] piperidine-2, 6-dione as a yellow oil residue or its enantiomer (61 mg,0.093mmol,99.51% yield, TFA) which was used without further purification.
Step 13:
to a solution of 3- [ 4-fluoro-1-oxo-5- [4- [ [ (2 s,5 s) -5- (piperazin-1-ylmethyl) tetrahydropyran-2-yl ] methyl ] piperazin-1-yl ] isoindolin-2-yl ] piperidine-2, 6-dione or its enantiomer (61 mg,0.093mmol,1 eq, TFA salt) and 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazole (35.75 mg,0.093mmol,1 eq) in DMSO (1 mL) was added DIEA (60.03 mg,0.46mmol,0.081mL,5 eq). The mixture was stirred at 100℃for 16 hours. LC-MS showed detection of the desired compound. The reaction mixture was partitioned between 2mL of water and 15mL (5 mL x 3) of DCM. The organic phase was separated, dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by preparative TLC (SiO 2, DCM: meoh=10:1). Purification was then carried out by means of preparative HPLC (column: unisil 3-100C18 Ultra 150*50mm*3um; mobile phase: [ water (FA) -ACN ]; B%:6% -36%,7 min). The compound 3- [ 4-fluoro-5- [4- [ [ (2 s,5 s) -5- [ [4- [6- [5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazol-3-yl ] pyrimidin-4-yl ] piperazin-1-yl ] methyl ] tetrahydropyran-2-yl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione or enantiomer thereof was obtained as a yellow solid (30 mg,0.033mmol,36.25% yield).
Step 14:
to a solution of 3- [ 4-fluoro-5- [4- [ [ (2 s,5 s) -5- [ [4- [6- [5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazol-3-yl ] pyrimidin-4-yl ] methyl ] tetrahydropyran-2-yl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione or its enantiomer (25 mg,0.028mmol,1 eq.) in DCM (1 mL) was added TFA (1.54 g,13.51mmol,1mL,481.38 eq.). The mixture was stirred at 20℃for 40 hours. LC-MS showed detection of the desired compound. The reaction mixture was concentrated in vacuo to give a residue. The residue was purified by preparative HPLC (column: unisil 3-100C18 Ultra150*50mm*3um; mobile phase: [ water (FA) -ACN ]; B%:1% -29%,7 min). The compound 3- [ 4-fluoro-5- [4- [ [ (2 s,5 s) -5- [ [4- [6- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] pyrimidin-4-yl ] methyl ] tetrahydropyran-2 yl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione, or an enantiomer thereof, was obtained as a yellow solid (5.9 mg,6.06umol,21.59% yield, 82.86% purity).
Exemplary synthesis of compound 306: compound 306 was prepared in a similar manner to compound 181 using the intermediate tert-butyl 3-fluoro-3- (hydroxymethyl) azetidine-1-carboxylate.
Exemplary synthesis of compound 307: compound 307 was prepared in a similar manner to compound 209.
Exemplary synthesis of compound 308: compound 308 was prepared in a similar manner to compound 181 using intermediate (4S) -4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] -2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester or an enantiomer thereof.
Step 1:
to a mixture of tert-butyl 2, 2-dimethyl-4-oxo-piperidine-1-carboxylate (10 g,43.99mmol,1 eq.) in ethanol (100 mL) was added sodium borohydride (9.18 g,242.62mmol,5.51 eq.) in small portions at 0 ℃. The mixture was stirred at 25℃for 2 hours. TLC showed the reaction was complete. Saturated aqueous ammonium chloride was added, ethanol was removed under reduced pressure, followed by extraction with ethyl acetate (20 ml×3). The combined organic phases were washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The crude product 4-hydroxy-2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester (10 g, crude) was obtained as a colorless oil.
/>
Step 2:
to a mixture of pyridin-4-ol (4.98 g,52.33mmol,1.2 eq.) and tert-butyl 4-hydroxy-2, 2-dimethyl-piperidine-1-carboxylate (10 g,43.61mmol,1 eq.) in tetrahydrofuran (100 mL) was added triphenylphosphine (17.16 g,65.41mmol,1.5 eq.) and diisopropyl azodicarboxylate (13.23 g,65.41mmol,12.7mL,1.5 eq.) at 0 ℃. The mixture was stirred at 25℃for 12 hours. LCMS showed detection of the desired m/z. The mixture was concentrated at 40 ℃ under reduced pressure. The residue was diluted with petroleum ether/ethyl acetate (v/v=3/1) (60 mL) and filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography (petroleum ether/ethyl acetate=10/1 to 3/1 to 1/1). The crude product, tert-butyl 2, 2-dimethyl-4- (4-pyridyloxy) piperidine-1-carboxylate (6.6 g, crude) was obtained as a colorless oil.
Step 3:
to a solution of tert-butyl 2, 2-dimethyl-4- (4-pyridyloxy) piperidine-1-carboxylate (6.00 g,19.6mmol,1 eq.) in acetic acid (20 mL) under nitrogen was added platinum oxide (1 g,4.40 mmol) and Pd/C (1 g,10% purity). The suspension was degassed under vacuum and treated with H 2 Purging several times. The mixture was stirred at 60℃under H 2 Stirring was carried out at (50 psi) for 48 hours. LCMS showed complete consumption of starting material. The reaction mixture was filtered, and the filtrate was concentrated. The residue was dissolved in water (50 mL) and neutralized to pH about 9 by 1N sodium hydroxide solution. The aqueous phase was extracted with ethyl acetate (30 ml×3), the combined organic phases were washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The crude product, tert-butyl 2, 2-dimethyl-4- (4-piperidinyloxy) piperidine-1-carboxylate (5.9 g, crude) was obtained as a colorless oil.
Step 4:
to a mixture of tert-butyl 2, 2-dimethyl-4- (4-piperidinyloxy) piperidine-1-carboxylate (5.2 g,16.64mmol,1 eq.) in tetrahydrofuran (100 mL) and water (50 mL) was added benzyl chloroformate (4.26 g,24.9mmol,3.6mL,1.5 eq.) and sodium bicarbonate (2.80 g,33.3mmol,2 eq.) at 0 ℃. The mixture was stirred at 25℃for 12 hours. LCMS showed the reaction was complete. The mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 ml×3). The combined organic phases were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (petroleum ether/ethyl acetate=20/1 to 10/1). The residue obtained was purified by preparative HPLC (column: phenomenex luna C (250X 70mm,10 um); mobile phase: [ water (FA) -ACN ];% B: 70% -100%,21 min). The residue was further separated by SFC (column: DAICEL CHIRALPAK AD (250 mm. Times.30 mm,10 um); mobile phase: [0.1% NH3H2OMEOH ]; B%:45% -45%,5.2 min). The compound (4S) -4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] -2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester or enantiomer thereof (1.25 g,2.80mmol,22.32% yield) was obtained as a colourless oil (rt=1.3 min). The compound (4R) -4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] -2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester or enantiomer thereof (1.25 g,2.80mmol,22.32% yield) was obtained as a colorless oil (rt=2.2 min).
Exemplary synthesis of compound 309: compound 309 is prepared in a similar manner to compound 209 using intermediate 4- [1- [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] -1-methyl-ethyl ] piperazine-1-carboxylic acid tert-butyl ester.
Step 1:
to a solution of 3, 4-difluoro-2-methyl-benzoic acid (50.00 g,290.48mmol,1 eq.) in methanol (500 mL) was added thionyl chloride (103.68 g,871.44mmol,63.2mL,3 eq.) dropwise. The mixture was stirred at 80℃for 12 hours. Thin layer chromatography showed the reaction was complete. The mixture was poured drop-wise into ice water (1L) at 0℃and the solid was filtered. The solid was dissolved in ethyl acetate (10 mL), filtered and concentrated to give methyl 3, 4-difluoro-2-methyl-benzoate (51.00 g,273.97mmol,94% yield) as a white solid.
Step 2:
to a mixture of methyl 3, 4-difluoro-2-methyl-benzoate (40.00 g,214.87mmol,1 eq.) in 1, 2-dichloroethane (400 mL) was added n-bromosuccinimide (57.37 g,322.31mmol,1.5 eq.) and benzoyl peroxide (520 mg,2.15mmol,0.01 eq.). The mixture was degassed and purged 3 times with nitrogen, then the mixture was stirred at 80 ℃ for 2 hours. Thin layer chromatography indicated that the reaction was complete. The mixture was cooled to 20 ℃, then filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether: ethyl acetate=1:0 to 20:1) to give methyl 2- (bromomethyl) -3, 4-difluoro-benzoate (51.00 g,192.42mmol,89% yield) as a colorless oil.
Step 3:
to a mixture of methyl 2- (bromomethyl) -3, 4-difluoro-benzoate (51.00 g,192.42mmol,1 eq.) and 3-aminopiperidine-2, 6-dione (33.25 g,202.04mmol,1.05 eq., hydrochloride) in N, N-dimethylformamide (600 mL) was added diisopropylethylamine (74.61 g,577.25mmol,100.5mL,3 eq.). The mixture was stirred at 40 ℃ for 1 hour, then heated to 110 ℃ for 12 hours. Thin layer chromatography indicated that the reaction was complete. The mixture was poured into water (800 mL) and filtered to obtain a solid. The solid was dissolved in ethyl acetate (500 mL), filtered and a solid was obtained. The compound 3- (4, 5-difluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (29.70 g,105.99mmol,55% yield) was obtained as a grey solid.
Step 4:
to a solution of 1-benzyloxycarbonyl piperidine-4-carboxylic acid (25 g,94.95mmol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester (17.69 g,94.95mmol,1 eq.) in N, N-dimethylformamide (500 mL) was added diisopropylethylamine (36.82 g,284.86mmol,49.62mL,3 eq.) and O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethylurea hexafluorophosphate (43.32 g,113.94mmol,1.2 eq.) at 0deg.C. The mixture was stirred at 25℃for 12 hours. LCMS showed the reaction was complete. The reaction mixture was quenched with water (1000 mL) at 20 ℃ and extracted with ethyl acetate (2000 ml×3), the combined organic layers were washed with brine (1500 ml×3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash chromatography on silica gel 80g/>Silica gel flash column, eluent: a gradient of 0% -100% ethyl acetate/petroleum ether, 60 mL/min) and trituration of the crude product with ethyl acetate (500 mL) at 20℃for 5 min. The compound tert-butyl 4- (1-benzyloxycarbonyl piperidine-4-carbonyl) piperazine-1-carboxylate (52 g,120.50mmol,63% yield, 100% purity) was obtained as a yellow oil.
Step 5:
to a mixture of zirconium chloride (17.28 g,74.16mmol,6.17mL,1.6 eq.) in tetrahydrofuran (500 mL) was added dropwise a solution of tert-butyl 4- (1-benzyloxycarbonyl piperidine-4-carbonyl) -piperazine-1-carboxylate (20 g,46.35mmol,1 eq.) in tetrahydrofuran (1L) at-78℃under nitrogen over a period of 30 minutes. Methyl magnesium bromide (3M, 77.25mL,5 eq.) was then added at-78deg.C and stirred for 30 min. The resulting mixture was warmed to 25 ℃ and stirred for 11 hours. LCMS showed the reaction was complete. The reaction was quenched by saturated aqueous ammonium chloride (1L) and stirred for 15 min. The aqueous phase was extracted with ethyl acetate (1L. Times.3). The combined organic phases were washed with brine (1 l×2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by semi-preparative reverse phase HPLC (column Phenomenex luna c, 250mm x 100mm x 10um; mobile phase: [ water (TFA) -ACN ];: B%:25% -50%,18 min). The compound 4- [1- (1-benzyloxycarbonyl-4-piperidinyl) -1-methyl-ethyl ] piperazine-1-carboxylic acid tert-butyl ester (23 g,51.62mmol,56% yield) was obtained as a colorless oil.
Step 6:
to a solution of 4- [1- (1-benzyloxycarbonyl-4-piperidinyl) -1-methyl-ethyl ] piperazine-1-carboxylic acid tert-butyl ester (5 g,11.22mmol,1 eq.) in trifluoroethanol (50 mL) under nitrogen was added palladium on carbon (2 g,10% purity). The suspension was degassed under vacuum and purged several times with hydrogen. The mixture was stirred at 30℃under hydrogen (50 psi) for 12 hours. LCMS showed the reaction was complete. The reaction mixture was filtered, and the filtrate was concentrated. Water (100 mL) was added and the mixture was extracted with ethyl acetate (200 mL. Times.3), and the aqueous phase was poured into aqueous sodium bicarbonate (100 mL) to adjust the pH to about 6-7, followed by extraction with ethyl acetate (200 mL. Times.3). The combined organic layers were washed with brine (150 ml×3), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The compound 4- [ 1-methyl-1- (4-piperidinyl) ethyl ] piperazine-1-carboxylic acid tert-butyl ester (3.9 g,9.17mmol,81.69% yield, trifluoroacetate salt) was obtained as a colorless oil.
Step 7:
to a solution of tert-butyl 4- [ 1-methyl-1- (4-piperidinyl) ethyl ] piperazine-1-carboxylate (1.14 g,2.68mmol,1.5 eq.) in dimethyl sulfoxide (5 mL) was added diisopropylethylamine (1.15 g,8.92mmol,1.55mL,5 eq.) and 3- (4, 5-difluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (500 mg,1.78mmol,1 eq.). The mixture was stirred at 120℃for 48 hours. LCMS showed the reaction was complete. Water (10 mL) was added and the mixture extracted with ethyl acetate (20 mL. Times.3). The combined organic layers were washed with brine (15 ml×3), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by recrystallisation from methanol (15 mL). The compound 4- [1- [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] -1-methyl-ethyl ] piperazine-1-carboxylic acid tert-butyl ester (280 mg,0.49mmol,27% yield) was obtained as a white solid.
Exemplary synthesis of compound 310: compound 310 was prepared in a similar manner to compound 309 using the intermediate 3- (4, 5-difluoro-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione and (4R) -4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] -2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester or an enantiomer thereof.
Exemplary synthesis of compound 311: compound 311 was prepared in an analogous manner to compound 181 starting from tert-butyl (3R) -3- (hydroxymethyl) pyrrolidine-1-carboxylate.
Exemplary synthesis of compound 312: compound 312 was prepared in a similar manner to compound 211.
Exemplary synthesis of compound 313: compound 313 is prepared in a similar manner to compound 310 using intermediate (4S) -4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] -2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester or an enantiomer thereof.
Exemplary synthesis of compound 314: compound 314 was prepared in a similar manner to compound 211 using the intermediate trans- [3- (4-piperidinyloxy) cyclobutyl ] methanol.
Exemplary synthesis of compound 315: compound 315 is prepared in a similar manner to compound 211 using intermediates to give 4- [3- [7- [2- (2, 6-dioxo-3-piperidinyl) -4-methoxy-1-oxo-isoindolin-5-yl ] -2, 7-diazaspiro [3.5] non-2-yl ] cyclobutoxy ] piperidine-1-carboxylic acid tert-butyl ester.
Step 1:
to 4- ((1 s,3 s) -3- (((trifluoromethyl) sulfonyl) oxy) cyclobutoxy) piperidine-1-carboxylic acid tert-butyl ester (404.98 mg,1.00mmol,1 eq.) and 3- [5- (2, 7-diazaspiro [ 3.5)]Non-7-yl) -4-methoxy-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (400 mg,1.00mmol,1 eq.) was added to a solution of MeCN (10 mL), DIEA (1.30 g,10.04mmol,1.75mL,10 eq.) and the mixture was stirred at 35℃for 48 h to give a yellow solution. LCMS showed the desired MS. TLC (dichloromethane: methanol=10:1) showed several new spots. Pouring the reaction mixture into H 2 O (20 mL). The mixture was extracted with DCM (20 mL x 3) and the organic phase was washed with brine (20 mL), over anhydrous Na 2 SO 4 Dried and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (0% to 10% methanol in dichloromethane) to give 4- [3- [7- [2- (2, 6-dioxo-3-piperidinyl) -4-methoxy-1-oxo-isoindolin-5-yl ] as a white solid]-2, 7-diazaspiro [3.5 ]]Non-2-yl]Cyclobutoxy radical]Tert-butyl piperidine-1-carboxylate (200 mg,187.18umol,18.65% yield, 61% purity).
Exemplary synthesis of compound 316: compound 316 was prepared in a similar manner to compound 211 using intermediate (4S) -4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] -2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester or an enantiomer thereof.
Exemplary synthesis of compound 317: compound 317 is prepared in an analogous manner to compound 209 starting with tert-butyl 5, 5-difluoro-2, 7-diazaspiro [3.5] nonane-2-carboxylate and using the intermediate (3S) -4- (2-chloroethyl) -3-methyl-piperazine-1-carboxylate.
Exemplary synthesis of compound 318: compound 318 was prepared in a similar manner to compound 211 using the intermediate tert-butyl 4- [ 1-methyl-1- (4-piperidinyl) ethyl ] piperazine-1-carboxylate.
Exemplary synthesis of compound 319: compound 319 is prepared in a similar manner to compound 211 using intermediate (4R) -4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] -2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester or an enantiomer thereof.
Exemplary synthesis of compound 320: compound 320 was prepared in a similar manner to compound 181 using the intermediate tert-butyl 4- [ 1-methyl-1- (4-piperidinyl) ethyl ] piperazine-1-carboxylate.
Exemplary synthesis of compound 321: compound 321 is prepared in a similar manner to compound 309 starting with tert-butyl (R) -3- (hydroxymethyl) pyrrolidine-1-carboxylate or an enantiomer thereof.
Exemplary synthesis of compound 322: compound 322 is prepared in a similar manner to compound 211 using the intermediate tert-butyl 4- [ 1-methyl-1- (4-piperidinyl) ethyl ] piperazine-1-carboxylate or an enantiomer thereof.
Exemplary synthesis of compound 323: compound 323 was prepared in a similar manner to compound 309 using the intermediate cis-4- [3- (trifluoromethylsulfonyloxy) cyclobutoxy ] piperidine-1-carboxylic acid tert-butyl ester.
Exemplary synthesis of compound 324: compound 324 was prepared in a similar manner to compound 211.
Exemplary synthesis of compound 325: compound 325 was prepared in a similar manner to compound 211 using intermediate (4S) -4- [ (1-benzyloxycarbonyl-4-piperidinyl) oxy ] -2, 2-dimethyl-piperidine-1-carboxylic acid tert-butyl ester or an enantiomer thereof.
Exemplary synthesis of compound 326: compound 326 was prepared in a similar manner to compound 209 using the intermediate tert-butyl 4- [ cis-3- (trifluoromethylsulfonyloxy) cyclobutoxy ] piperidine-1-carboxylate.
Exemplary synthesis of compound 327: compound 327 is prepared in a similar manner to compound 181 starting from tert-butyl (R) -3- (hydroxymethyl) pyrrolidine-1-carboxylate.
Exemplary synthesis of compound 328: compound 328 was prepared in a similar manner to compound 211 starting from tert-butyl (R) -3- (hydroxymethyl) pyrrolidine-1-carboxylate.
Exemplary synthesis of compound 329: compound 329 was prepared in a similar manner to compound 290 using the intermediate 3- (5-bromo-4-methoxy-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione.
Exemplary synthesis of compound 330: compound 330 was prepared in a similar manner to compound 309 starting from tert-butyl (S) -3- (hydroxymethyl) pyrrolidine-1-carboxylate.
Exemplary synthesis of compound 331: compound 331 was prepared in a similar manner to compound 211 using the intermediate tert-butyl 4- [ 1-methyl-1- (4-piperidinyl) ethyl ] piperazine-1-carboxylate.
Exemplary synthesis of compound 332: compound 332 was prepared in a similar manner to compound 211 starting from benzyl 5, 5-difluoro-2, 7-diazaspiro [3.5] nonane-7-carboxylate.
Exemplary synthesis of compound 333: compound 333 was prepared in a similar manner to compound 209 using the intermediate 3- (5-bromo-4-methyl-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione.
Step 1:
to a stirred solution of methyl 4-bromo-2, 3-dimethyl-benzoate (1 g,4.11mmol,1 eq.) in DCE (10 mL) under nitrogen was added NBS (878.58 mg,4.94mmol,1.2 eq.) followed by 2- [ (E) - (1-cyano-1-methyl-ethyl) azo]2-methyl-propionitrile (33.77 mg,205.68umol,0.05 eq) and the resulting mixture was vigorously stirred at 90℃for 2 hours. TLC showed completion. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (petroleum ether) to give 4-bromo-2- (bromomethyl) -3-methyl-benzoic acid as a yellow oilMethyl ester (1.4 g, crude).
Step 2:
to a mixture of methyl 4-bromo-2- (bromomethyl) -3-methyl-benzoate (1.4 g,4.35mmol,1 eq.) and 3-aminopiperidine-2, 6-dione (858.75 mg,5.22mmol,1.2 eq., HCl) in DMF (10 mL) was added DIEA (2.81 g,21.74mmol,3.79mL,5 eq.) at 20 ℃ in one portion. The mixture was stirred at 85 ℃ for 10 hours. LCMS showed a new peak with the desired MS. The reaction mixture was concentrated in vacuo and the resulting mixture was purified with MeCN (20 mL) and H 2 O (20 mL) was ground at 20deg.C. The crude material was filtered and the solid concentrated in vacuo to give 3- (5-bromo-4-methyl-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (800 mg,2.37mmol,54.57% yield) as a dark grey solid.
Exemplary synthesis of compound 334: compound 334 was prepared in a similar manner to compound 209 using intermediate tert-butyl 4- (((1 r,3 r) -3-formylcyclobutyl) methyl) piperazine-1-carboxylate.
Step 1:
at-40 ℃ at N 2 Downward methoxymethyl (triphenyl) phosphonium; to a mixture of chloride (80.26 g,234.15mmol,1.5 eq.) in THF (50 mL) was added NaHMDS (1M, 234.15mL,1.5 eq.). The mixture was stirred at 0deg.C for 30 min, then cooled to-40deg.C, and a solution of methyl 3-oxocyclobutanoate (20 g,156.10mmol,1 eq.) in THF (20 mL) was added. The reaction mixture was stirred at 25 ℃ for 16 hours. TLC showed the reaction was complete. Pouring the residue into saturated NH 4 Cl (100 mL). The aqueous phase was extracted with MTBE (3X 100 mL). The combined organics were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography220nm,0% -2% (2 min) MTBE/petroleum ether, 2% (10 min) MTBE/petroleum ether to give methyl 3- (methoxymethylene) cyclobutanecarboxylate (9.4 g,60.19mmol,38.56% yield) as a colorless oil.
Step 2:
to a solution of methyl 3- (methoxymethylene) cyclobutanecarboxylate (9.3 g,59.55mmol,1 eq.) in DCM (50 mL) was added TFA (13.58 g,119.09mmol,8.82mL,2 eq.) and H 2 O (5 mL). The resulting mixture was stirred at 25℃for 2 hours. TLC showed the reaction was complete. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (220 nm,80g,0% -10% (10 min) MTBE/petroleum ether, 10% (30 min) MTBE/petroleum ether) to give methyl trans-3-formylcyclobutane carboxylate (1.7 g,11.96mmol,20.08% yield) and methyl cis-3-formylcyclobutane carboxylate (3.4 g,23.92mmol,40.17% yield) as colorless oils.
Step 3:
to a mixture of trans-3-formylcyclobutane methyl formate (1.5 g,10.55mmol,1 eq.) and piperazine-1-carboxylic acid tert-butyl ester (3.93 g,21.10mmol,4.08mL,2 eq.) in DCM (30 mL) and HOAc (1 mL) at 25℃was added NaBH at once 3 CN (1.33 g,21.10mmol,2 eq.). The mixture was stirred at 25℃for 1 hour. TLC and LCMS indicated that the reaction was complete. The residue was poured into water (30 mL). The aqueous phase was extracted with ethyl acetate (30 ml x 3). The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (0% -38% (25 min) ethyl acetateEthyl acetate/petroleum ether, 38% (25 min) ethyl acetate/petroleum ether) to give 4- [ (3-methoxycarbonylcyclobutyl) methyl as a colorless oil]Piperazine-1-carboxylic acid tert-butyl ester (1.5 g,4.80mmol,45.49% yield).
Step 4:
at 0 ℃ at N 2 Downward 4- [ (3-methoxycarbonylcyclobutyl) methyl group]To a mixture of tert-butyl piperazine-1-carboxylate (1.5 g,4.80mmol,1 eq.) in THF (30 mL) was added LiAlH 4 (364.43 mg,9.60mmol,27.09mL,2 eq.). The mixture was stirred at 0 ℃ for 1 hour. TLC showed the reaction was complete. Pouring the residue into H 2 O(1mL)、NaOH(1mL,15%)、H 2 O (3 mL). The aqueous phase was extracted with ethyl acetate (40 ml x 3). The combined organic phases were washed with brine (40 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0% -100% petroleum ether/petroleum ether) to give 4- [ [3- (hydroxymethyl) cyclobutyl ] as a yellow oil]Methyl group]Piperazine-1-carboxylic acid tert-butyl ester (1.2 g,4.01mmol,83.51% yield, 95% purity).
Step 5:
at 25 ℃ at N 2 Downward 4- [ [3- (hydroxymethyl) cyclobutyl ]]Methyl group]To a mixture of tert-butyl piperazine-1-carboxylate (400 mg,1.41mmol,1 eq.) in DCM (10 mL) was added DMP (1.19 g,2.81mmol,870.89uL,2 eq.) in one portion. The mixture was stirred at 25 ℃ for 1 hour to give a yellow solution. TLC (petroleum ether: ethyl acetate=0:1) showed complete consumption of starting material. Pouring the residue into NaHCO 3 To adjust ph=7-8, and Na is added 2 SO 3 (20 mL). The aqueous phase was extracted with DCM (30 ml x 3). The combined organic phases were washed with brine (30 ml x 2), dried over anhydrous Na 2 SO 4 Drying, filtering andconcentrated in vacuo to give tert-butyl 4- (((1 r,3 r) -3-formylcyclobutyl) methyl) piperazine-1-carboxylate (400 mg, crude) as a yellow oil.
Exemplary synthesis of compound 335: compound 335 was prepared in a similar manner to compound 309 using the intermediate tert-butyl 4- [ cis-3- (trifluoromethylsulfonyloxy) cyclobutoxy ] piperidine-1-carboxylate.
Exemplary synthesis of compound 336: compound 336 was prepared in a similar manner to compound 317.
Exemplary synthesis of compound 337: compound 337 was prepared in a similar manner to compounds 209 and 309 using the intermediate tert-butyl 3-fluoro-3- (4-piperidinylmethyl) azetidine-1-carboxylate.
Step 1:
at-70 ℃ at N 2 n-BuLi (2.5M, 10.31mL,1.2 eq.) was added dropwise to a solution of 4-methylpyridine (2 g,21.48mmol,2.11mL,1 eq.) in THF (20 mL). The mixture was stirred at-70 ℃ for 3 hours to give an orange solution, followed by dropwise addition of tert-butyl 3-oxo-azetidine-1-carboxylate (3.68 g,21.48mmol,1 eq.) in THF (10 mL), and the solution was stirred at 20 ℃ for 2 hours to give a pale yellow solution. TLC and LCMS showed the reaction was complete. The residue was poured into water (20 mL). The aqueous phase was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (40 g,0% -60% (30 min) ethyl acetate/petroleum ether, 60% (15 min) ethyl acetate/petroleum ether) to give tert-butyl 3-hydroxy-3- (4-pyridylmethyl) azetidine-1-carboxylate (3.31 g,12.52mmol,58.31% yield) as a yellow gum.
Step 2:
at-40 ℃ at N 2 DAST (2.42 g,15.03mmol,1.99mL,1.2 eq.) was added dropwise to a mixture of tert-butyl 3-hydroxy-3- (4-pyridylmethyl) azetidine-1-carboxylate (3.31 g,12.52mmol,1 eq.) in DCM (100 mL). The reaction was then heated to 20 ℃ and stirred for 30 minutes to give an orange solution. TLC (petroleum ether: ethyl acetate=1:1, rf=0.74) showed no starting material and a new spot. Cool the reaction to 0deg.C and use NaHCO 3 The aqueous solution was quenched to ph=7-8. The aqueous phase was extracted with DCM (10 ml x 3). The combined organic layers were washed with brine (10 ml x 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (40 g,0% -18% (15 min) ethyl acetate/petroleum ether, 18% (15 min) ethyl acetate/petroleum ether) to give tert-butyl 3-fluoro-3- (4-pyridylmethyl) azetidine-1-carboxylate (2.33 g,8.75mmol,69.87% yield) as a pale yellow solid.
Step 3:
to a solution of tert-butyl 3-fluoro-3- (4-pyridylmethyl) azetidine-1-carboxylate (2.33 g,8.75mmol,1 eq.) in EtOH (20 mL) and HOAc (525.39 mg,8.75mmol,500.37uL,1 eq.) was added PtO at 25deg.C 2 (298.01 mg,1.31mmol,0.15 eq.). The mixture is heated to 70℃under H 2 Stirring was carried out at (50 psi) for 24 hours. TLC showed the reaction was complete. The suspension was filtered through a celite pad and the pad was washed with EtOH (100 ml×3). The combined filtrates were concentrated to dryness to give tert-butyl 3-fluoro-3- (4-piperidinylmethyl) azetidine-1-carboxylate (2.8 g, crude) as a colorless oil.
Exemplary synthesis of compound 338: compound 338 was prepared in a similar manner to compound 211 using the intermediate 3- (6-chloropyrimidin-4-yl) -5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [3,4-c ] pyridine.
Exemplary synthesis of compound 339: compound 339 was prepared in a similar manner to compound 209.
Exemplary synthesis of compound 340: compound 340 was prepared in a similar manner to compound 211.
Exemplary synthesis of compound 341: compound 341 was prepared in a similar manner to compound 211.
Exemplary synthesis of compound 342: compound 342 was prepared in an analogous manner to compound 297 using the intermediates 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione and (3 r,4 s) -4- (2, 6-dioxopiperidin-3-yl) -4-fluoro-1-oxo-isoindolin-5-yl) -3-fluoropiperidine-1-carboxylic acid tert-butyl ester.
Exemplary synthesis of compound 343: compound 343 was prepared in a similar manner to compound 209.
Exemplary synthesis of compound 344: compound 344 was prepared in a similar manner to compound 211 using intermediate 4- ((1 s,3 s) -3- (((trifluoromethyl) sulfonyl) oxy) cyclobutoxy) piperidine-1-carboxylic acid tert-butyl ester.
Exemplary synthesis of compound 345: compound 345 was prepared in a similar manner to compound 211 using the intermediate tert-butyl 3-fluoro-3- (4-piperidinylmethyl) azetidine-1-carboxylate.
Exemplary synthesis of compound 346: compound 346 was prepared in a similar manner to compound 209.
Exemplary synthesis of compound 347: compound 347 was prepared in a similar manner to compound 211 using intermediate 2- ((1 r,3 r) -3- (piperidin-4-yloxy) cyclobutyl) -2, 7-diazaspiro [3.5] nonane-7-carboxylic acid benzyl ester.
Step 1:
to 2, 7-diazaspiro [3.5]]To a solution of t-butyl nonane-2-carboxylate (3 g,13.26mmol,1 eq.) and TEA (1.61 g,15.91mmol,2.21mL,1.2 eq.) in THF (20 mL) was added CbzCl (2.71 g,15.91mmol,2.26mL,1.2 eq.) and the mixture stirred at 0deg.C for 2 hours to give a yellow solution. TLC (Petroleum ether: ethyl)Ethyl acrylate=3:1, rf=0.17) shows no starting material and shows new spots. Pouring the obtained product into H 2 0 (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The organic phase was washed with brine (20 mL), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether) to give 2, 7-diazaspiro [3.5 ] as a colorless oil]Nonane-2, 7-dicarboxylic acid O7-benzyl ester O2-tert-butyl ester (3.5 g,9.71mmol,73.25% yield).
Step 2:
to 2, 7-diazaspiro [3.5 ]]To a solution of nonane-2, 7-dicarboxylic acid O7-benzyl ester O2-tert-butyl ester (3.5 g,9.71mmol,1 eq.) in DCM (5 mL) was added TFA (6.16 g,54.03mmol,4mL,5.56 eq.). After the addition, the reaction mixture was stirred at 25 ℃ for 1 hour. TLC (petroleum ether: ethyl acetate=3:1) showed a new spot and LCMS showed the desired MS. Addition of NaHCO 3 The aqueous solution was used to adjust ph=7-8. The aqueous phase was extracted with DCM (15 ml x 3). The organic layer was concentrated under reduced pressure to give 2, 7-diazaspiro [3.5 ] as a colorless oil]Nonane-7-carboxylic acid benzyl ester (2 g,7.68mmol,79.12% yield).
Step 3:
to 2, 7-diazaspiro [3.5 ]]Benzyl nonane-7-carboxylate (1.4 g,5.38mmol,1 eq.) and 4- [3- (trifluoromethylsulfonyloxy) cyclobutoxy ]To a solution of tert-butyl piperidine-1-carboxylate (2.82 g,6.99mmol,1.3 eq.) in MeCN (15 mL) was added DIEA (2.09 g,16.13mmol,2.81mL,3 eq.) and the mixture was stirred at 60℃for 12 h to give a yellow solution. TLC (petroleum ether: ethyl acetate=0:1, rf=0.01) showed several new spots. TLC (dichloromethane: methanol=10:1, rf=0.6) showed several new spots. The obtained product is processedPouring the mixture into H 2 O (10 mL). The mixture was extracted with DCM (20 ml x 3). The organic phase was washed with brine (20 mL), dried over anhydrous Na 2 SO 4 Drying and concentrating in vacuo gave a residue. The residue was purified by silica gel column chromatography (0% to 100% ethyl acetate/petroleum ether) and the residue was purified by silica gel column chromatography (0% to 10% methanol/dichloromethane) to give 2- [3- [ (1-tert-butoxycarbonyl-4-piperidinyl) oxy ] as a pale yellow gum]Cyclobutyl group]-2, 7-diazaspiro [3.5 ]]Nonane-7-carboxylic acid benzyl ester (1.67 g,2.41mmol,44.74% yield, 74% purity).
Step 4:
to 2- [3- [ (1-tert-butoxycarbonyl-4-piperidinyl) oxy]Cyclobutyl group]-2, 7-diazaspiro [3.5 ]]To a solution of benzyl nonane-7-carboxylate (1.3 g,2.53mmol,1 eq.) in DCM (5 mL) was added TFA (3.08 g,27.01mmol,2mL,10.67 eq.). After the addition, the reaction mixture was stirred at 25 ℃ for 1 hour. TLC (dichloromethane: methanol=10:1) showed a new spot and LCMS showed the desired MS. Addition of NaHCO 3 The aqueous solution was used to adjust ph=7-8. The aqueous phase was extracted with DCM (15 ml x 3). The organic layer was concentrated under reduced pressure to give 2- ((1 r,3 r) -3- (piperidin-4-yloxy) cyclobutyl) -2, 7-diazaspiro [3.5 ] as a white solid]Nonane-7-carboxylic acid benzyl ester (93mg, 2.25mmol,88.86% yield).
Exemplary synthesis of compound 348: compound 348 was prepared in a similar manner to compound 234.
Exemplary synthesis of compound 349: compound 349 was prepared in a similar manner to compounds 181 and 211.
Exemplary synthesis of compound 350: compound 350 was prepared in a similar manner to compound 309.
Exemplary synthesis of compound 351: compound 351 was prepared in a similar manner to compound 209 using intermediate ((1 r,3 r) -3- (piperidin-4-yloxy) cyclobutyl) methanol.
Exemplary synthesis of compound 352: compound 352 was prepared in a similar manner to compound 234.
Exemplary synthesis of compound 353: compound 353 was prepared in a similar manner to compound 309.
Exemplary synthesis of compound 354:
step 1:
at 0 ℃ at N 2 Downward [4- (4-piperidinyloxy) cyclohexyl]To a mixture of methanol (30 g,140.64mmol,1 eq.) in DCM (300 mL) was added CbzCl (31.19 g,182.83mmol,25.99mL,1.3 eq.) and TEA (42.69 g,421.91mmol,58.72mL,3 eq.). After addition, the reaction mixture was stirred at 25 ℃ for 1 hour to give a pale yellow suspension. TLC (petroleum ether: ethyl acetate=1:1, rf=0.3) showed a new spot for the reaction. The reaction was purified by adding H 2 O (200 mL) and extracted with ethyl acetate (3X 100 mL). The combined organic phases were washed with water, with Na 2 SO 4 Dried and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (0% -100% ethyl acetate/petroleum ether) to give 4- [4- (hydroxymethyl) cyclohexyloxy ] as a colorless oil]Benzyl piperidine-1-carboxylate (37 g,106.49mmol,75.72% yield).
Step 2:
to 4- [4- (hydroxymethyl) cyclohexyloxy]Benzyl piperidine-1-carboxylate (37 g,106.49mmol,1 eq.) was added to a solution of DMP (55.50 g,130.85mmol,40.51mL,1.23 eq.) in DCM (200 mL) and the mixture stirred at 25℃for 2 h to give a yellow solution. TLC (petroleum ether: ethyl acetate=1:1, rf=0.6) showed a new spot for the reaction. The reaction mixture was cooled to 0deg.C and purified by addition of saturated NaHCO 3 To a pH of about 8 and extracted with DCM (100 ml x 3). The combined organic layers were taken up in saturated Na 2 SO 3 (100 ml x 2) and brine (60 ml x 2), washed with Na 2 SO 4 Dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (petroleum ether/ethyl acetate=100/1, 1/1) to give benzyl 4- (4-formylcyclohexyloxy) piperidine-1-carboxylate (30 g,85.98mmol,80.74% yield, 99% purity) as a colorless oil.
Step 3:
to a mixture of benzyl 4- (4-formylcyclohexyloxy) piperidine-1-carboxylate (30 g,86.85mmol,1 eq.) in MeOH (200 mL) at 25℃was added TOsOH (747.77 mg,4.34mmol,0.05 eq.) and trimethoxymethane (46.08 g,434.24mmol,47.60mL,5 eq.) in one portion. The mixture was stirred at 25℃for 2 hours. TLC (petroleum ether: ethyl acetate=3:1, rf=0.5) showed a new spot for the reaction. Passing the reaction through H 2 O (100 mL) was quenched and extracted with ethyl acetate (3 x 200 mL). The combined organic phases were washed with water, with Na 2 SO 4 Dried and concentrated in vacuo to give a residue. The residue was purified by silica gel column chromatography (0% -30% ethyl acetate/petroleum ether) to give 4- [4- (dimethoxymethyl) cyclohexyloxy ] as a colorless oil]Benzyl piperidine-1-carboxylate (31 g,75.22mmol,86.62% yield, 95% purity).
Step 4:
at 25 ℃ at H 2 (15 PSI) for 16 hours to 4- [4- (dimethoxymethyl) cyclohexyloxy)]To a mixture of benzyl piperidine-1-carboxylate (31 g,79.18mmol,1 eq.) in EtOH (200 mL) was added Pd/C (8 g,79.18mmol,10% purity, 1 eq.). TLC (petroleum ether: ethyl acetate=3:1) showed a new spot. The residue was filtered and concentrated in vacuo to give 4- [4- (dimethoxymethyl) cyclohexyloxy as a white solid Base group]Piperidine (18.6 g,72.27mmol,91.27% yield).
Step 5:
to a solution of 4- [4- (dimethoxymethyl) cyclohexyloxy ] piperidine (500 mg,1.94mmol,1 eq.) in DMSO (5 mL) was added 4-bromo-2-fluoro-pyridine (341.90 mg,1.94mmol,1 eq.) and DIEA (502.16 mg,3.89mmol,676.77uL,2 eq.). After the addition, the reaction solution was stirred at 90 ℃ for 1 hour. LCMS showed the desired MS and TLC (petroleum ether: ethyl acetate=5:1) showed a new spot. After cooling, the reaction solution was diluted with ethyl acetate (10 mL) and washed with water (3×10 mL). The organic layer was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether) to give 4-bromo-2- [4- [4- (dimethoxymethyl) cyclohexyloxy ] -1-piperidinyl ] pyridine (680 mg,1.60mmol,82.14% yield, 97% purity) as a yellow solid.
Step 6:
to a solution of 5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazole (5 g,18.36mmol,1 eq.) in THF (80 mL) was added Pin 2 B 2 (11.66 g,45.90mmol,2.5 eq.) and (1Z, 5Z) -cycloocta-1, 5-diene; 2, 4-dimethyl-BLAH bicyclo [1.1.0]Butane (1.22 g,1.84mmol,0.1 eq.). After addition, the reaction mixture was taken up in N at 25 ℃ 2 Stirred for 12 hours. LCMS showed a peak with the desired MS and TLC (petroleum ether: ethyl acetate=3:1) showed the reaction was complete. The reaction mixture was combined with the individual reactants for work-up. The combined reaction mixtures were filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 23% ethyl acetate/petroleum ether) to give 5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-3- (4, 5-tetramethylene) as a pale yellow solid1,3, 2-Dioxolanyl-2-yl) indazole (9.5 g,17.17mmol,93.54% yield, 72% purity). Based on the combined reaction, the average yield was 77.9%.
Step 7:
to 4-bromo-2- [4- [4- (dimethoxymethyl) cyclohexyloxy]-1-piperidinyl group]Pyridine (415.11 mg,1.00mmol,1 eq.) and 5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-3- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) indazole (400 mg,1.00mmol,1 eq.) in dioxane (5 mL) and H 2 Pd (dppf) Cl was added to the solution in O (1 mL) 2 (73.48 mg,100.43umol,0.1 eq.) and Na 2 CO 3 (319.33 mg,3.01mmol,3 eq.). After addition, the reaction mixture was taken up in N at 80 ℃ 2 Stirred for 16 hours. LCMS showed a new peak with the desired MS and TLC (petroleum ether: ethyl acetate=3:1) showed a new spot. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether) to give 3- [2- [4- [4- (dimethoxymethyl) cyclohexyloxy ] as a brown oil ]-1-piperidinyl group]-4-pyridinyl]-5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazole (560 mg,787.07umol,78.37% yield, 85% purity).
Step 8:
to 3- [2- [4- [4- (dimethoxymethyl) cyclohexyloxy)]-1-piperidinyl group]-4-pyridinyl]To a solution of 5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazole (400 mg,661.40umol,1 eq.) in THF (4 mL) was added HCl (2 m,4mL,12.10 eq.). After the addition, the reaction solution was stirred at 25 ℃ for 10 minutes. TLC (petroleum ether: ethyl acetate=3:1) showed a new spot. NaHCO 3 Added to the reaction mixture to adjust the ph=8-9. The mixture was extracted with ethyl acetate (10 mL) and the organic layer was washed with water (3X 15 mL). The mixture was concentrated under reduced pressure to give 4- [ [1- [4- [5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazol-3-yl ] as a yellow solid]-2-pyridyl group]-4-piperidinyl]Oxy group]Cyclohexane formaldehyde (300 mg, crude).
Step 9:
to 4- [ [1- [4- [5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazol-3-yl ]]-2-pyridyl group]-4-piperidinyl]Oxy group]Cyclohexane Formaldehyde (160 mg,286.37umol,1 eq) and 3- (4-fluoro-1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (99.19 mg,286.37umol,1 eq) were added borane: 2-methylpyridine (91.89 mg,859.12umol,3 eq) in a mixture of MeOH (10 mL) and AcOH (2 mL) and the mixture was stirred at 25℃for 12 hours. LCMS showed the desired MS and TLC (dichloromethane: methanol=10:1) showed a new spot. NaHCO 3 To the reaction mixture was added to adjust ph=8-9, the mixture was extracted with ethyl acetate (10 mL), and the extract was washed with water (3×15 mL). The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 10% methanol in dichloromethane) to give 3- [ 4-fluoro-5- [4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazol-3-yl ] as a white solid]-2-pyridyl group]-4-piperidinyl]Oxy group]Cyclohexyl group]Methyl group]Piperazin-1-yl]-1-oxo-isoindolin-2-yl]Piperidine-2, 6-dione (200 mg,195.71umol,68.34% yield, 87% purity).
Step 10:
to a mixture of 3- [ 4-fluoro-5- [4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] oxy ] cyclohexyl ] methyl ] piperazin-1-yl ] -1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (150 mg,168.72umol,1 eq.) in DCM (5 mL) was added TFA (3.08 g,27.01mmol,2mL,160.11 eq.) and the mixture stirred at 25 ℃ for 12 hours. LCMS showed a new peak with the desired MS. The mixture was concentrated and purified by preparative HPLC (column: phenomenex C18 75X 30mm 3um; mobile phase: [ water (FA) -ACN ]; B%:6% -36%,25 min) to afford 3- [ 4-fluoro-5- [4- [ [4- [ [1- [4- [5- (1-methylcyclopropoxy) -1H-indazol-3-yl ] -2-pyridinyl ] -4-piperidinyl ] oxy ] cyclohexyl ] methyl ] piperazin-1-oxo-isoindolin-2-yl ] piperidine-2, 6-dione (36.1 mg,44.69umol,26.49% yield, 99.65% purity) as a white solid.
Exemplary synthesis of compound 355: compound 355 is prepared in a similar manner to compound 209.
Exemplary synthesis of compound 356: compound 356 was prepared in a similar manner to compound 211 using the intermediate tert-butyl 3- (4-piperidinyloxy) azetidine-1-carboxylate.
Exemplary synthesis of compound 357: compound 357 is prepared in a similar manner to compound 211.
Exemplary synthesis of compound 358: compound 358 was prepared in a similar manner to compound 78.
Exemplary synthesis of compound 359: compound 359 was prepared in a similar manner to compounds 181 and 211.
Exemplary synthesis of compound 360: compound 360 was prepared in a similar manner to compounds 211 and 234.
Exemplary synthesis of compound 361: compound 361 was prepared in a similar manner to compound 309 using intermediate 4- (((1 r,4 r) -4- ((2, 2-dimethylpiperazin-1-yl) methyl) cyclohexyl) oxy) piperidine-1-carboxylic acid tert-butyl ester.
Step 1:
to a mixture of 4-hydroxycyclohexanecarboxylic acid (3.1 g,21.50mmol,1 eq.) in dimethylformamide (50 mL) was added O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethylurea (12.26 g,32.25mmol,1.5 eq.) in one portion at 0deg.C, and the mixture was stirred for 30 minutes. Then 3, 3-dimethylpiperazine-1-carboxylic acid tert-butyl ester (5.07 g,23.65mmol,1.1 eq.) and diisopropylethylamine (5.56 g,43.01mmol,7.49mL,2 eq.) were added at 0deg.C. The mixture was warmed to 25 ℃ and stirred for 16 hours. LCMS showed the reaction was complete. Water (100 mL) was added and the mixture was extracted with ethyl acetate (50 mL. Times.3). The combined organic phases were washed first with 0.5mol of hydrochloric acid (70 mL. Times.1), then with saturated sodium bicarbonate solution (50 mL. Times.1), then with water (50 mL) and finally with brine (50 mL. Times.2). The resulting organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The compound 4- (4-hydroxycyclohexanecarbonyl) -3, 3-dimethyl-piperazine-1-carboxylic acid tert-butyl ester (11.3 g, crude product) was obtained as a black oil.
Step 2:
to a mixture of tert-butyl 4- (4-hydroxycyclohexanecarbonyl) -3, 3-dimethyl-piperazine-1-carboxylate (7.32 g,21.50mmol,1 eq.) was added hydrochloric acid/dioxane (4 m,70ml,13.02 eq.) in one portion at 25 ℃. The mixture was stirred at 25℃for 1 hour. LCMS showed the reaction was complete. The reaction mixture was concentrated under reduced pressure to remove the solvent. The compound (2, 2-dimethylpiperazin-1-yl) - (4-hydroxycyclohexyl) methanone (11.2 g, crude) was obtained as a black oil.
Step 3:
to a mixture of (2, 2-dimethylpiperazin-1-yl) - (4-hydroxycyclohexyl) methanone (5.17 g,21.51mmol,1 eq.) and sodium bicarbonate (9.04 g,107.56mmol,4.18mL,5 eq.) in tetrahydrofuran (10 mL) and water (5 mL) was added benzyl chloroformate (4.04 g,23.66mmol,3.36mL,1.1 eq.) at 0℃in one portion. The mixture was stirred at 25℃for 16 hours. LCMS showed the reaction was complete. The mixture was diluted with water (100 mL), followed by extraction with ethyl acetate (50 ml×3). The combined organic phases were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (petroleum ether/ethyl acetate=5/1 to 1/2). Benzyl 4- (4-hydroxycyclohexanecarbonyl) -3, 3-dimethyl-piperazine-1-carboxylate (4.21 g,11.24mmol,52% yield) was obtained as a yellow oil.
Step 4:
to a mixture of benzyl 4- (4-hydroxycyclohexanecarbonyl) -3, 3-dimethyl-piperazine-1-carboxylate (4.21 g,11.24mmol,1 eq.) in tetrahydrofuran (40 mL) was added borane dimethyl sulfide complex (10 m,6.75mL,6 eq.) in one portion at 0 ℃. The mixture was stirred at 65℃for 16 hours. LCMS showed the reaction was complete. The solution was quenched with 1.0N hydrochloric acid solution (about 100 mL). The impurity was extracted with ethyl acetate (40 mL. Times.3). The aqueous phase was adjusted to ph=7-8 with saturated sodium bicarbonate solution (about 100 mL). The mixture was extracted with ethyl acetate (80 ml×3), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give benzyl 4- [ (4-hydroxycyclohexyl) methyl ] -3, 3-dimethyl-piperazine-1-carboxylate (2.95 g,8.18mmol,72% yield) as a yellow oil.
Step 5:
to a mixture of benzyl 4- [ (4-hydroxycyclohexyl) methyl ] -3, 3-dimethyl-piperazine-1-carboxylate (2.9 g,8.04mmol,1 eq.) in tetrahydrofuran (30 mL) was added trimethylchlorosilane (961.38 mg,8.85mmol,1.12mL,1.1 eq.) and trimethylamine (1.22 g,12.07mmol,1.68mL,1.5 eq.) at 0 ℃ in one portion. The mixture was stirred at 25℃for 1 hour. Thin layer chromatography showed the reaction was complete. The mixture was concentrated at 40 ℃ under reduced pressure. The residue was dissolved in dichloromethane (30 mL) and 1- (2, 2-trifluoroacetyl) piperidin-4-one (1.57 g,8.04mmol,1 eq.) was added and the mixture was cooled to-70 ℃. Triethylsilane (1.87 g,16.09mmol,2.57mL,2 eq.) was added followed by a slow addition of trimethylsilane triflate (1.79 g,8.04mmol,1.45mL,1 eq.). The mixture was stirred at 25℃for 16 hours. LCMS showed the reaction was complete. Saturated aqueous sodium bicarbonate (100 mL) was added to the mixture. The aqueous phase was extracted with ethyl acetate (100 mL. Times.3). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated in vacuo. Benzyl 3, 3-dimethyl-4- [ [4- [ [1- (2, 2-trifluoroacetyl) -4-piperidinyl ] oxy ] cyclohexyl ] methyl ] piperazine-1-carboxylate (4.6 g, crude) was obtained as a yellow oil.
Step 6:
to a solution of benzyl 3, 3-dimethyl-4- [ [4- [ [1- (2, 2-trifluoroacetyl) -4-piperidinyl ] oxy ] cyclohexyl ] methyl ] piperazine-1-carboxylate (2.98 g,5.52mmol,1 eq.) in methanol (30 mL) was added potassium carbonate (2.29 g,16.57mmol,3 eq.) in one portion at 25 ℃. The mixture was stirred at 25℃for 2 hours. Thin layer chromatography showed the reaction was complete. The reaction mixture was filtered, and the filtrate was diluted with water (100 mL) and extracted with dichloromethane (30 ml×3). The combined organic phases were washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Benzyl 3, 3-dimethyl-4- [ [4- (4-piperidinyloxy) cyclohexyl ] methyl ] piperazine-1-carboxylate (2 g,4.51mmol,81% yield) was obtained as a yellow oil, which was used in the next step without further purification.
Step 7:
to a mixture of benzyl 3, 3-dimethyl-4- [ [4- (4-piperidinyloxy) cyclohexyl ] methyl ] piperazine-1-carboxylate (2 g,4.51mmol,1 eq.) and 4-dimethylaminopyridine (55 mg,0.45mmol,0.1 eq.) in dichloromethane (20 mL) was added triethylamine (912 mg,9.02mmol,1.3mL,2 eq.) and di-tert-butyl dicarbonate (1.48 g,6.76mmol,1.5mL,1.5 eq.) at 0 ℃ in one portion. The mixture was stirred at 25℃for 16 hours. LCMS showed the reaction was complete. 30mL of saturated aqueous ammonium chloride solution was added. The mixture was then extracted with ethyl acetate (50 ml×3). The combined organic phases were washed with 30mL of saturated sodium bicarbonate solution, 30mL of water and 50mL of brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (petroleum ether/ethyl acetate=10/1 to 3/1). 4- [ [4- [ (1-tert-butoxycarbonyl-4-piperidinyl) oxy ] cyclohexyl ] methyl ] -3, 3-dimethyl-piperazine-1-carboxylic acid benzyl ester (1.7 g,3.13mmol,69% yield) was obtained as a yellow oil.
Step 8:
to a solution of benzyl 4- [ [4- [ (1-tert-butoxycarbonyl-4-piperidinyl) oxy ] cyclohexyl ] methyl ] -3, 3-dimethyl-piperazine-1-carboxylate (1.7 g,3.13mmol,1 eq.) in ethyl acetate (20 mL) at 25 ℃ palladium on carbon (170 mg,3.13mmol,10% purity, 1 eq.) was added in one portion. The mixture was stirred at 25 ℃ under a hydrogen atmosphere for 3 hours. LCMS showed the reaction was complete. The reaction mixture was filtered. Tert-butyl 4- (((1 r,4 r) -4- ((2, 2-dimethylpiperazin-1-yl) methyl) cyclohexyl) oxy) piperidine-1-carboxylate (1.28 g, crude) was obtained as an oil, which was used in the next step without further purification.
Exemplary synthesis of compound 362: compound 362 was prepared in a similar manner to compound 309.
Exemplary synthesis of compound 363: compound 363 was prepared in a similar manner to compound 211.
Exemplary synthesis of compound 364: compound 364 was prepared in a similar manner to compound 211.
Exemplary synthesis of compound 365: compound 365 was prepared in a similar manner to compound 354.
Exemplary synthesis of compound 366: compound 366 was prepared in a similar manner to compound 211.
Exemplary synthesis of compound 367: compound 367 was prepared in a similar manner to compounds 354 and 211.
Exemplary synthesis of compound 368: compound 368 was prepared in a similar manner to compound 235.
Exemplary synthesis of compound 369: compound 369 is prepared in a similar manner to compound 211.
Exemplary synthesis of compound 370: compound 370 was prepared in a similar manner to compound 239.
Exemplary synthesis of compound 371: compound 371 was prepared in a similar manner to compound 78 using the intermediate 3-iodo-5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [4,3-b ] pyridine.
/>
Step 1:
at 20 ℃ at N 2 Pd (dba) was added in one portion to a mixture of 6-bromo-2-methyl-3-nitro-pyridine (6 g,27.65mmol,1 eq.) and 1-methylcyclopropanol (1.99 g,27.65mmol,1 eq.) in toluene (10 mL) 2 (317.95 mg,552.94umol,0.02 eq.) Cs 2 CO 3 (10.81 g,33.18mmol,1.2 eq.) and BINAP (1.03 g,1.66mmol,0.06 eq.). The mixture was heated to 110 ℃ and stirred for 3 hours. LCMS showed the desired MS. The mixture was cooled to 20 ℃. The residue was poured into water (10 mL). The mixture was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (petroleum ether) to give 2-methyl-6- (1-methylcyclopropoxy) -3-nitro-pyridine (3.41 g,16.38mmol,59.24% yield) as a yellow oil.
Step 2:
at 20 ℃ at N 2 Pd/C (500 mg,18.73mmol,10% purity, 1 eq.) and ammonium formate (14.17 g,224.77mmol,12 eq.) were added in one portion to a mixture of 2-methyl-6- (1-methylcyclopropoxy) -3-nitro-pyridine (3.90 g,18.73mmol,1 eq.) in EtOH (10 mL). The mixture was stirred at 20 ℃ for 1 hour to give a black solution. TLC showed the reaction was complete. The mixture was filtered through a pad of silica gel, the pad was washed with EtOAc (3×200 mL), and the organics were concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% ethyl acetate/petroleum ether) to give 2-methyl-6- (1-methylcyclopropoxy) pyridin-3-amine (3.5 g, crude) as a red oil.
Step 3:
at 0 ℃ at N 2 under To 2-methyl-6- (1-methylcyclopropoxy) pyridin-3-amine (3.54 g,19.86mmol,1 eq.) and Et 3 N (5.02 g,49.65mmol,6.91mL,2.5 eq.) Ac was added in one portion to a mixture of DCM (10 mL) 2 O (4.06 g,39.72mmol,3.72mL,2 eq.). The mixture was stirred at 0 ℃ for 30 minutes, then heated to 20 ℃ and stirred for 1 hour. TLC showed the reaction was complete and LCMS showed a peak with the desired MS. The reaction was purified by the reaction with saturated NaHCO 3 (30 mL) ph=7-8, and the mixture was quenched with CH 2 Cl 2 (3X 50 mL) extraction. The combined organic phases were washed with brine (3X 50 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 20% -40% ethyl acetate/petroleum ether) to give N- [ 2-methyl-6- (1-methylcyclopropoxy) -3-pyridinyl as a red oil]Acetamide (4.2 g,19.07mmol,96.00% yield).
Step 4:
to N- [ 2-methyl-6- (1-methylcyclopropoxy) -3-pyridinyl at 20 DEG C]To a solution of acetamide (4.20 g,19.07mmol,1 eq.) in toluene (10 mL) was added KOAc (2.81 g,28.60mmol,1.5 eq.) and Ac 2 O (8.95 g,87.71mmol,8.22mL,4.6 eq.) the solution was heated to 80℃followed by dropwise addition of isoamyl nitrite (8.93 g,76.27mmol,10.26mL,4 eq.). The mixture was stirred at 80℃for 16 hours. TLC and LCMS showed the reaction was complete. The reaction was filtered, the resulting solid was washed with EtOAc (70 mL) and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (100-200 mesh silica gel, 0% -10% ethyl acetate/petroleum ether) to give 1- [5- (1-methylcyclopropoxy) pyrazolo [4,3-b ] as a yellow oil ]Pyridin-1-yl]Ethanone (2.9 g,12.54mmol,65.77% yield).
Step 5:
to 1- [5- (1-methylcyclopropoxy) pyrazolo [4,3-b ] at 20 DEG C]Pyridin-1-yl]To a solution of ethanone (2.90 g,12.54mmol,1 eq.) in MeOH (50 mL) was added ammonia (7M, 1.79mL,1 eq.) (NH 3 (g) /MeOH). The mixture was stirred at 20 ℃ for 30 minutes to give a red solution. LCMS showed the reaction was complete. The solution was concentrated in vacuo to give 5- (1-methylcyclopropoxy) -1H-pyrazolo [4,3-b as a yellow solid]Pyridine (2.7 g, crude).
Step 6:
to 5- (1-methylcyclopropoxy) -1H-pyrazolo [4,3-b]To a solution of pyridine (1.3 g,6.87mmol,1 eq.) in DMF (15 mL) was added I 2 (3.49 g,13.74mmol,2.77mL,2 eq.) and KOH (1.16 g,20.61mmol,3 eq). After the addition, the reaction mixture was stirred at 25 ℃ for 1 hour. TLC (petroleum ether: ethyl acetate=5:1) showed a major new spot, and LCMS showed a peak with the desired MS. The reaction mixture was purified by adding saturated Na 2 S 2 O 3 (30 mL) was quenched and extracted with ethyl acetate (2X 30 mL). The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 15% ethyl acetate/petroleum ether) to give 3-iodo-5- (1-methylcyclopropoxy) -1H-pyrazolo [4,3-b ] as a colorless oil ]Pyridine (1.8 g,5.71mmol,83.14% yield).
Step 7:
at 0 ℃ at N 2 Downward 3-iodo-5- (1-methylcyclopropoxy) -1H-pyrazolo [4,3-b]To a mixture of pyridine (1.8 g,5.71mmol,1 eq.) in THF (20 mL) was added NaH (274.19 mg,6.85mmol,60% purity, 1.2 eq.) in one portion. The mixture was stirred at 0deg.C for 30 min, followed by the addition of [ chloro (diphenyl) methyl ]]Benzene (1.91 g,6.85mmol,1.2 eq.) and the solution was stirred at 25℃for 2 hours. TLC and LCMS showed the reaction was complete. The residue was poured into water (20 mL). The mixture was extracted with ethyl acetate (20 ml x 3). The combined organic phases were washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (40 g,0% -5% (10 min) ethyl acetate/petroleum ether) to give 3-iodo-5- (1-methylcyclopropoxy) -1-trityl-pyrazolo [4,3-b ] as a white solid]Pyridine (2.7 g,4.84mmol,84.79% yield).
Exemplary synthesis of compound 372: compound 372 was prepared in a similar manner to compound 209 using the intermediate benzyl 4- (2-azaspiro [3.5] non-7-yloxymethyl) piperidine-1-carboxylate.
Step 1:
at 0 ℃ at N 2 Downward 7-oxo-2-azaspiro [3.5] ]To a solution of tert-butyl nonane-2-carboxylate (1.9 g,7.94mmol,1 eq.) in MeOH (20 mL) was added NaBH 4 (0.62 g,16.39mmol,2.06 eq.). The mixture was stirred at 0 ℃ for 1 hour to give a colorless solution. TLC (petroleum ether: ethyl acetate=3:1, rf=0.22) showed that the reaction was complete. The mixture was poured into HCl (10 ml,2 m) and extracted with DCM (20 ml x 3). The organic layer was washed with brine (20 ml x 2), dried over anhydrous Na 2 SO 4 Drying, filtering and concentrating under vacuum to give 7-hydroxy-2-azaspiro [3.5 ] as a white solid]Nonane-2-carboxylic acid tert-butyl ester (1.8 g,7.46mmol,93.95% yield).
Step 2:
at 40 ℃ at N 2 Downward t-Buona (1.19 g,12.43mmol,2 eq.) and 7-hydroxy-2-azaspiro [3.5 ]]To a mixture of tert-butyl nonane-2-carboxylate (1.5 g,6.22mmol,1 eq.) in DMSO (10 mL) was added 4- (chloromethyl) pyridine (872.23 mg,6.84mmol,1.1 eq.) in one portion. The mixture was stirred at 80℃for 10 hours. LCMS showed a peak with the desired MS. The mixture was cooled to 20 ℃. The residue was poured into water (10 mL) and stirred for 10 min. The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under vacuum. The crude product was purified by reverse phase HPLC (column: welch Xtimate C18X 50mm 10um; mobile phase: [ Water (FA) -ACN) ]The method comprises the steps of carrying out a first treatment on the surface of the B%:0% -38%,27 min) to give 7- (4-pyridylmethoxy) -2-azaspiro [3.5 ] as a yellow oil]Nonane-2-carboxylic acid tert-butyl ester (520 mg,1.56mmol,25.17% yield).
Step 3:
to 7- (4-pyridylmethoxy) -2-azaspiro [3.5 ] under N2]To a solution of tert-butyl nonane-2-carboxylate (520 mg,1.56mmol,1 eq.) in MeOH (10 mL) was added PtO2 (100 mg,440.37umol,2.82e-1 eq.) and HOAc (93.93 mg,1.56mmol,89.46uL,1 eq.). The suspension was degassed under vacuum and treated with H 2 Purging several times. The mixture is heated to 70℃under H 2 Stirring for 10 hours at (50 psi). LCMS showed a peak with the desired MS. The suspension was filtered through a pad of celite and the filter cake was washed with EtOAc (20 ml×3) and the filtrate was concentrated to give 7- (4-piperidinylmethoxy) -2-azaspiro [3.5 ] as a yellow oil]Nonane-2-carboxylic acid tert-butyl ester (1.13 g, crude, HOAc).
Step 4:
to 7- (4-piperidinylmethoxy) -2-azaspiro [3.5 ] at 0deg.C]Nonane-2-carboxylic acid tert-butyl ester (600 mg,1.77mmol,1 eq.) and NaHCO3 (1.49 g,17.73mmol,689.40uL,10 eq.) in H 2 To a solution of O (10 mL) and MeCN (10 mL) in a mixed solvent was added CbzCl (453.59 mg,2.66mmol,377.99uL,1.5 eq). The reaction mixture was stirred at 0 ℃ for 1 hour. TLC showed the reaction was complete. The residue was poured into water (10 mL). The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (0% -15% ethyl acetate/petroleum ether) to give 7- [ (1-benzyloxycarbonyl-4-piperidinyl) methoxy group as a colorless oil]-2-azaspiro [3.5 ]]Nonane-2-carboxylic acid tert-butyl ester (79mg, 1.67mmol,94.30% yield).
Step 5:
at 25 ℃ at N 2 Downward 7- [ (1-benzyloxycarbonyl-4-piperidinyl) methoxy]-2-azaspiro [3.5 ]]Nonane-2-carboxylic acid tert-butyl ester (86To a mixture of 0mg,1.82mmol,1 eq.) in DCM (10 mL) was added TFA (2.40 g,21.08mmol,1.56mL,11.59 eq.) in one portion. The mixture was stirred at 25℃for 30 minutes. LCMS showed the reaction was complete. Pouring the residue into NaHCO 3 (10 mL) to adjust ph=7-8. The aqueous phase was extracted with ethyl acetate (10 ml x 3). The combined organic phases were washed with brine (10 ml x 2), dried over anhydrous Na 2 SO 4 Drying, filtration and concentration under vacuum gives 4- (2-azaspiro [3.5 ] as a white solid]Benzyl nono-7-yloxymethyl) piperidine-1-carboxylate (694 mg, crude).
Exemplary synthesis of compound 373: compound 373 was prepared in a similar manner to compound 219.
Exemplary synthesis of compound 374: compound 374 was prepared in a similar manner to compound 209.
Exemplary synthesis of compound 375: compound 375 was prepared in a similar manner to compound 78.
Exemplary synthesis of compound 376: compound 376 was prepared in a similar manner to compound 181.
Exemplary synthesis of compound 377: compound 377 was prepared in a similar manner to compound 181 using the intermediate 3- (6-chloropyridazin-4-yl) -5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazole.
Step 1:
to 5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-3- (4, 5-tetramethyl-1, 3, 2-dioxapentaborane-2-yl) indazole (180 mg,451.92umol,1 eq.) and 5-bromo-3-chloro-pyridazine (87.41 mg,451.92umol,1 eq.) in dioxane (10 mL) and H 2 Pd (dppf) Cl was added to the solution in O (2 mL) 2 (33.07 mg,45.19umol,0.1 eq.) and Na 2 CO 3 (143.70 mg,1.36mmol,3 eq.). After addition, the reaction mixture was taken up at 100℃under N 2 Stirred for 16 hours. LCMS showed a peak with the desired MS and TLC (petroleum ether: ethyl acetate=3:1) showed a peak of oneNew blobs. After cooling, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (0% to 20% ethyl acetate/petroleum ether) to give 3- (6-chloropyridazin-4-yl) -5- (1-methylcyclopropoxy) -1-tetrahydropyran-2-yl-indazole (150 mg,323.50umol,71.58% yield, 83% purity) as a white solid.
Exemplary synthesis of compound 378: compound 378 is prepared in a similar manner to compound 309.
Exemplary synthesis of compound 379: compound 379 was prepared in a similar manner to compound 354.
Exemplary synthesis of compound 380: compound 380 was prepared in a similar manner to compounds 210 and 211.
Exemplary synthesis of compound 381: compound 381 is prepared in a similar manner to compound 234.
Exemplary synthesis of compound 382: compound 382 was prepared in a similar manner to compound 211.
Exemplary synthesis of compound 383: compound 383 was prepared in a similar manner to compound 354 using the intermediate 4- [ [1- [2- (2, 6-dioxo-3-piperidinyl) -4-fluoro-1-oxo-isoindolin-5-yl ] -4-piperidinyl ] oxy ] cyclohexanecarboaldehyde.
Exemplary synthesis of compound 384: compound 384 was prepared in a similar manner to compounds 354 and 211.
Detailed description of the preferred embodiments
One aspect of the present disclosure relates to a heterobifunctional compound having the chemical structure:
PTM―L―CLM,
or a pharmaceutically acceptable salt or solvate thereof,
wherein:
(a) The CLM is a small molecule E3 ubiquitin ligase binding moiety that binds cereblon E3 ubiquitin ligase and is represented by the following chemical structure:
Wherein:
w is CH 2 、C=O、SO 2 Or NH;
each X is independently selected from the group consisting of absent, O and S;
z is absent, O or S;
g is H or unsubstituted or substituted straight or branched alkyl;
each Q 1 、Q 2 、Q 3 And Q 4 N, CH or CR independently;
a is H or unsubstituted or substituted straight or branched alkyl;
n is an integer from 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched alkyl, optionally substituted alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L; and is also provided with
R 'and R' are independently selected from the group consisting of a bond, H, and optionally substituted alkyl;
represents a single bond or a double bond; and is also provided with
Represents a bond that may be stereotactic ((R) or (S)) or non-stereotactic;
(b) The PTM is a small molecule leucine rich repeat kinase 2 (LRRK 2) targeting moiety that binds to LRRK2 or a mutant form thereof, represented by the following chemical structure:
/>
wherein:
R 1 is isopropyl, tert-butyl,Wherein->Is the point of attachment to the oxygen atom of the PTM;
R 2 is hydrogen, F, cl, OH, C1-C3 alkyl or C1-C3 fluoroalkyl;
X 6 and X 7 Each independently is CH or N;
X 1 、X 2 、X 3 、X 4 and X 5 Each independently is N or CH, when CH, optionally R 2 Substitution;
X 8 is CH or N;
is an optionally substituted 3-10 membered cycloalkyl, heterocycloalkyl, bicycloalkyl, spirocycloalkyl or spiroheterocycloalkyl containing 1-4 (e.g., 1, 2, 3 or 4) heteroatoms selected from N, O and S (e.g., optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents); and is also provided with
The PTM isRepresents a point of attachment to the L; and is also provided with
(c) The L is a chemical linker group that covalently couples the CLM to the PTM.
In any aspect or embodiment described herein, the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
In any aspect or embodiment described herein, the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
One aspect of the present disclosure relates to a heterobifunctional compound having the chemical structure:
PTM―L―CLM,
or a pharmaceutically acceptable salt or solvate thereof,
wherein:
(a) The CLM is a small molecule E3 ubiquitin ligase binding moiety that binds cereblon E3 ubiquitin ligase and is represented by the following chemical structure:
wherein:
w is CH 2 、C=O、SO 2 Or NH;
each X is independently selected from the group consisting of absent, O and S;
z is absent, O or S;
G is H or unsubstituted or substituted straight or branched alkyl;
each Q 1 、Q 2 、Q 3 And Q 4 N, CH or CR independently;
a is H or unsubstituted or substituted straight or branched alkyl;
n is an integer from 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched alkyl, optionally substituted alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L; and is also provided with
R 'and R' are independently selected from the group consisting of a bond, H, and optionally substituted alkyl;
represents a single bond or a double bond; and is also provided with
Represents a bond that may be stereotactic ((R) or (S)) or non-stereotactic;
(b) The PTM is a small molecule leucine rich repeat kinase 2 (LRRK 2) targeting moiety that binds to LRRK2 or a mutant form thereof, represented by the following chemical structure:
/>
R 1 is isopropyl, tert-butyl,Wherein->Is the point of attachment to the oxygen atom of the PTM;
R 2 is hydrogen, F, cl, OH, C1-C3 alkyl or C1-C3 fluoroalkyl;
X 6 and X 7 Each independently is CH or N;
X 1 、X 2 、X 3 、X 4 and X 5 Each independently is N or CH, when CH, optionally R 2 Substitution;
X 8 is CH or N;
is an optionally substituted 3-10 membered cycloalkyl, heterocycloalkyl, bicycloalkyl, spirocycloalkyl or spiroheterocycloalkyl containing 1-4 (e.g., 1, 2, 3 or 4) heteroatoms selected from N, O and S (e.g., optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents); and is also provided with
The PTM isRepresents a point of attachment to the L; and is also provided with
(c) The L is a bond or a chemical linker group that covalently couples the CLM to the PTM.
In any aspect or embodiment described herein, the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
In any aspect or embodiment described herein, the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
In any aspect or embodiment described herein, the CLM is represented by the following chemical structure:
in any aspect or embodiment described herein, the compound is represented by a chemical structure selected from the group consisting of:
/>
wherein:
X 2 c, CH or N;
Z 1 is a bond, carbon shared with a cyclic group of L, or nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
In any aspect or embodiment described herein, the compound is represented by a chemical structure selected from the group consisting of:
wherein:
X 2 c, CH or N;
Z 1 is a bond, carbon shared with a cyclic group of L, or nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
In any aspect or embodiment described herein, the compound is represented by the following chemical structure:
in any aspect or embodiment described herein, the compound is represented by the following chemical structure:
/>
in any aspect or embodiment described herein,
in any aspect or embodiment described herein, each R is selected from H, O, OH, NH, NH 2 、-Cl、-F、CN、CF 3 Optionally substituted straight-chain or branched C 1-3 Alkyl, optionally substituted straight or branched C 1-3 An alkoxy group.
In any aspect or embodiment described herein, each R is selected from H, O, OH, NH, NH 2 、-Cl、-F、-CN、CF 3 Methyl, methoxy and ethoxy.
In any aspect or embodiment described herein, one or more of the following:
(a)is->
Wherein: r is R 3 Is H or methyl; r is R 3a Is H, halogen (e.g., cl or F) or methyl; r is R 4 Is H or A
A base; r is R 5 Is H or methyl;representing the point of attachment to the PTM; and->Represents the point of attachment to L and wherein +.>
There is no time for the existence of the non-woven fabric,through atoms of N or CH of cyclic groups, R 3 、R 4 Or R is 5 To L;
(b)R 2 is H or F; or (b)
(c) A combination thereof.
In any aspect or embodiment described herein,selected from the group consisting of:
in any aspect or embodiment described herein,/>
wherein said heterocycloalkyl is attached to said L or said PTM through an atom of said cyclic group or a substituent thereof.
In any aspect or embodiment described herein, the PTM is represented by the following chemical structure:
/>
/>
/>
/>
/>
/>
/>
wherein the method comprises the steps ofRepresents the attachment site of the L.
In any aspect or embodiment described herein, the L comprises a chemical structural unit represented by the formula: - (A) L ) q -, wherein:
-(A L ) q -is a group attached to the CLM and the PTM;
q is an integer greater than or equal to 1;
each A L Independently selected from CR L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 C.ident.C, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 0-6R L1 And/or R L2 Aryl substituted with groups and optionally with 1-6R L1 And/or R L2 A group-substituted heteroaryl group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
In any aspect or embodiment described herein, the L comprises optionally substituted C 1 -C 50 Alkyl (e.g., C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 7 、C 8 、C 9 、C 10 、C 11 、C 12 、C 13 、C 14 、C 15 、C 16 、C 17 、C 18 、C 19 、C 20 、C 21 、C 22 、C 23 、C 24 、C 25 、C 26 、C 27 、C 28 、C 29 、C 30 、C 31 、C 32 、C 33 、C 34 、C 35 、C 36 、C 37 、C 38 、C 39 、C 40 、C 41 、C 42 、C 43 、C 44 、C 45 、C 46 、C 47 、C 48 、C 49 Or C 50 Alkyl), wherein:
each carbon is optionally replaced by: CR (computed radiography) L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 C.ident.C, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 1-6R L1 And/or R L2 Aryl substituted by radicals or optionally substituted by 1-6R L1 And/or R L2 A group-substituted heteroaryl group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
In any aspect or embodiment described herein, the L is selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
the chemical linker group is optionally substituted with halogen;
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Each m, n, o, p, q and r of the L is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20.
In any aspect or embodiment described herein, one or more of the following:
(a) The CLM is represented by:
/>
wherein: the +.>Represents a point of attachment to the L; and N is a nitrogen atom shared with the L;
(b) The PTM is represented by:
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein the PTM is covalently linked to the L through an atom of the heterocycloalkyl a or a substituent thereof;
(c) The L is a linker group (L) selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Representing a point of attachment to said CLM or said PTM; or (b)
(d) A combination thereof.
In any aspect or embodiment described herein, one or more of the following: the PTM is a PTM selected from the group consisting of compounds 52-288, the CLM is a CLM selected from the group consisting of compounds 52-288, and the L is an L selected from the group consisting of compounds 52-288.
In any aspect or embodiment described herein, one or more of the following:
g is H or unsubstituted or substituted straight-chain or branched C 1-6 An alkyl group;
a is H or unsubstituted or substituted straight-chain or branched C 1-6 An alkyl group;
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched C 1-6 Alkyl, optionally substituted C 1-6 Alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L;
r 'and R' are independently selected from the group consisting of bond, H and optionally substituted C 1-6 An alkyl group; and is also provided with
Is an optionally substituted 3-10 membered cycloalkyl, an optionally substituted 3-10 membered heterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered bicycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered spirocycloalkyl, or an optionally substituted 3-10 membered spiroheterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, wherein said heteroatoms are independently selected from N, O and S.
In any aspect or embodiment described herein, the PTM has the following chemical structure:
in any aspect or embodiment described herein, the compound has the following chemical structure:
wherein:
X 2 c, CH or N;
Z 1 is a bond, carbon shared with a cyclic group of L, or nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
In any aspect or embodiment described herein, the compound is represented by the following chemical structure:
in any aspect or embodiment described herein, the compound is represented by the following chemical structure:
in any aspect or embodiment described herein, one or more of the following:
(a)is->/>
Wherein:
R 3 is H or methyl;
R 3a is H, halogen or methyl;
R 4 is H or methyl;
R 5 is H or methyl;
representing a point of connection to the PTM; and is also provided with
Represents the point of attachment to said L and when +.>In the absence of said-> Through atoms of N or CH, R of said cyclic group 3 Or R is 4 Is connected to the L;
(b)R 2 is H or F; or (b)
(c) A combination thereof.
In any aspect or embodiment described herein, whereinSelected from the group consisting of: wherein said heterocycloalkyl is attached to said L or said PTM through an atom of said cyclic group or a substituent thereof.
In any aspect or embodiment described herein, the PTM is represented by the following chemical structure:
/>
/>
wherein->Represents the attachment site of the L.
In any aspect or embodiment described herein, the L is selected from the group consisting of:
/>
wherein:
the chemical linker group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen and methyl (preferably independently selected from halogen);
C is a carbon atom covalently linked or shared with said CLM or said PTM;
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Each m, n, and o of the L is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.
In any aspect or embodiment described herein, one or more of the following:
(a) The CLM is represented by:
/>
/>
wherein:
said ULMRepresents a point of attachment to the L;
c is a carbon atom shared with the L; and is also provided with
N is a nitrogen atom shared with the L;
(b) The PTM is represented by:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein the PTM is covalently linked to the L through an atom of the heterocycloalkyl a or a substituent thereof;
(c) The L is a linker group (L) selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
n is a nitrogen atom covalently linked or shared with said CLM or said PTM;
c is a carbon atom covalently linked or shared with said CLM or said PTM; and is also provided with
Representing a point of attachment to said CLM or said PTM; or (b)
(d) A combination thereof.
In any aspect or embodiment described herein, one or more of the following: the PTM is a PTM selected from the compounds of table 1, the CLM is a CLM selected from the compounds of table 1, and the L is L selected from the compounds of table 1.
In any aspect or embodiment described herein, the compound is represented by the following chemical structure:
wherein:
n is 0 or 1;
r is H, OH, -Cl, -F or Br;
Z 1 is nitrogen or carbon shared with the cyclic group of L;
X 2 is C or N;
X 4 is CH or N (preferably N);
R 1 is that(preferably->) Wherein->Is the point of attachment to the oxygen of the PTM;
R 2 is H, -Cl or-F;
the method comprises the following steps: />
(preferably-> ) Wherein:
R 3 is H, methyl or ethyl;
R 4 is H, methyl or ethyl;
representing said->A connection point with the PTM; and is also provided with
Represents the junction of said PTM with said L and when +.>In the absence of said->May be attached to the L through an atom of the cyclic group (e.g., carbon or nitrogen);
l is represented by the following chemical structure:
/>
wherein: />
m and n are integers independently selected from 0, 1, 2 or 3 (preferably 1); and is also provided with
The L is optionally substituted with 0, 1, 2 or 3 (preferably 0 or 1) groups selected from: -Cl, -F and C 1-3 Alkyl (e.g., methyl or ethyl).
In any aspect or embodiment described herein, X 2 Is C and X 4 Is N.
Another aspect of the present disclosure relates to a composition comprising an effective amount of a bifunctional compound of the present disclosure and a pharmaceutically acceptable carrier.
In any aspect or embodiment described herein, the composition further comprises an additional bioactive agent.
In any aspect or embodiment described herein, the additional bioactive agent is an anti-inflammatory agent, a chemotherapeutic agent, or an immunomodulatory agent.
Another aspect of the present disclosure relates to a composition comprising a pharmaceutically acceptable carrier and an effective amount of at least one compound of the present disclosure for treating a disease, disorder, or symptom of causal relationship with LRRK2 in a subject, wherein the composition is effective to treat or ameliorate the disease, disorder, or at least one symptom of the disease or disorder.
One aspect of the present disclosure relates to a method for treating a disease, disorder, or symptom having a causal relationship with LRRK2, wherein the method comprises administering to a subject in need thereof a composition comprising a pharmaceutically acceptable carrier and an effective amount of at least one compound of the present disclosure, wherein the composition is effective to treat or ameliorate the disease, disorder, or at least one symptom of the disease or disorder.
Another aspect of the present disclosure relates to a method of treating or preventing a disease, disorder, or condition associated with LRRK2, the method comprising providing to a patient in need thereof an effective amount of a compound as described herein or a composition comprising the compound and administering to the patient an effective amount of a compound as described herein or a composition comprising the compound, wherein the compound or composition is effective to treat or ameliorate the disease, disorder, or at least one symptom of the disease or disorder.
In any aspect or embodiment described herein, the disease or disorder is idiopathic Parkinson's Disease (PD), LRRK2 mutation-related PD, primary tauopathy, lewy body dementia, crohn's disease, leprosy, neuroinflammation, progressive supranuclear palsy, pick's disease, ftdttau, TDP-43 frontotemporal dementia, TDP-43ALS, c9orf ALS, huntington's disease, spinocerebellar ataxia (SCAs) 1, 2, 3, 6, 7 and 17, dentate nuclear pallidoluffing (DRPLA) or kennedy's disease.
Protein level control
The present specification also provides methods for controlling intracellular protein levels. The method is based on the use of a compound as described herein such that degradation of the target protein LRRK2 in vivo will result in a reduced amount of target protein in the biological system, preferably to provide a specific therapeutic benefit.
The following examples are provided to aid in the description of the present disclosure, but should not be construed to limit the disclosure in any way.
In certain embodiments, the present description provides the following exemplary LRRK2 degrading bifunctional molecules (compounds or compounds 52-384 of table 1), including salts, polymorphs, analogs, derivatives and deuterated forms thereof.
Examples for testing LRRK2 degradation driven by exemplary heterobifunctional compounds designed to target LRRK2 Sex determination
This assay measures the degradation of wild type and G2019S LRRK2 (labeled with a HiBit tag on the C-terminus of the protein) in HEK293 cells, which is expressed by mammalian expression vectors driven by ubiquitin promoters. Dose responses for each compound were repeated on two separate days, each day on three separate plates.
Plasmid preparation. The transfection mixture was assembled as follows and incubated for 30 min at room temperature. 5.25mL of Opti-MEM (without additives) was mixed by flicking with 1. Mu.g/. Mu.L of 17. Mu.L firefly luciferase plasmid and 1. Mu.g/. Mu.L of 158. Mu.L of WT plasmid DNA (175. Mu.g total DNA) in a 15mL tube. In a new 15mL tube, 5.25mL of OptiMEM was mixed by flicking with 1. Mu.g/. Mu.L of 17. Mu.L firefly luciferase plasmid and 1. Mu.g/. Mu.L of 158. Mu. L G2019S plasmid DNA (175. Mu.g total DNA). X-trememeGene HP was thoroughly mixed using a vortex. Next, 175 μl was added to each tube and flicked to mix. Both tubes were incubated for 30 minutes at room temperature.
HEK293 cells (obtained from ATCC; ATCC CRL-1573) were harvested with trypsin when the transfection mixture was incubated. Once the cells were detached, the cells were resuspended in 12mL optmem+5% FBS and transferred to a 50mL tube. The cells were thoroughly mixed and counted. Cells were plated in two 250mL conical tubes at 0.71X 10 using OptiMEM+5% FBS 6 Individual cells/mL dilution (70 mL). One tube is labeled "WT" and the other tube is labeled "G2019S". WT and G2019S transfection mixtures were added drop-wise to the corresponding 250mL tubes. The tube was mixed first by pipetting followed by vortexing. The tubes were incubated at room temperature for at least 5 minutes.
The tubes were vortexed before dispensing and after every third plate. 70 microliter of cells were each dispensed into seven plates with either WT or G2019 SDNA. Three panels each were tested for compound panel one (preparation as described below) and three panels each were tested for compound panel two (preparation as described below). The first plate in each set was used as the "master" plate and was not used for testing compounds. Each plate was incubated in a fume hood for 10 minutes, after which it was placed in a 37 ℃ incubator for 24 hours.
Preparation of compounds and assay plates. Two compound plates were prepared using 96-well polypropylene plates. The compound was made to 10mM and diluted to 1mM in 30. Mu.L. Each dose-response curve included wells of DMSO (as negative control and for normalization) and wells of 0.5 μm compound 4 (as positive control). In addition to the seven test compounds, each plate also included a dose response of compound 4. The compound plates were spun down at 1200rpm for 2 minutes.
The two compound plates were then mixed and 2 μl diluted in the middle plate containing 248 μl Opti-Mem in each well. Next, 10 μl of the diluted compound from the intermediate plate was added to each test plate (three WT plates and three G2019S plates per compound plate, total 12 assay plates). Plates were incubated at 37℃for 24 hours.
All assay plates and all Nano-Glo dual fluorescent reporter assay system components (except DLR substrate) were equilibrated to room temperature. Next, the luciferase buffer was mixed with a lyophilized amber vial until completely dissolved, and 75 μl of the luciferase mixture was added to each well of each assay plate. The assay plates were incubated for 10 minutes at room temperature with shaking for at least 5 minutes, followed by reading on a plate reader.
Color plate and analytical data. 1mL of DLR substrate and 1mL of LgBiT protein were added to Stop and Glo buffer, and 75. Mu.L of the mixture was added to each well of each plate. An optically clear seal was added to each plate and each plate incubated for 20 minutes with shaking for at least 10 minutes followed by reading on a plate reader.
As mentioned above, the plates were run in triplicate and the assay was repeated twice (6 replicates total for each exemplary compound). Firefly luciferase was examined for cell number and viability per cell, and Nanoluc was examined for LRRK2-HiBit quantification.
The (HiBit/luciferase) ratio was determined to be 1000 and the data normalized to median% DMSO. Curve fitting was performed for each individual plate. The data for the exemplary compounds of table 1 below are shown in the columns G2019SDC50 (nM), G2019SDmax (%), WT DC50 (nM), and WT Dmax (%) in table 2 below.
For testing by design for targetingExemplary heterobifunctional compounds for LRRK 2-driven LRRK2 degradation Sex determination
This assay measures the degradation of LRRK2 in cells where the C-terminal (3') of the endogenous gene has been tagged with HiBit sequences in HEK293 cells. These cells also express firefly luciferase, which is expressed by the cytomegalovirus promoter and introduced into HiBit-tagged cells and stably expressed. UtilizesDual luciferase reporter assay system (Promega TM ,Madison,WI)。
Day 1-preparation of compounds and assay plates. Two sets of plates were prepared: one set of triplicates was used for HiBit assays in white 384 well plates and one set of triplicates was used for Alamar Blue cell viability assays in black 384 well plates. Briefly, the growth medium (DMEM+Glutamax-10% fetal bovine serum-1% penicillin-streptomycin) in both T128 flasks was aspirated from the flasks. Cells were washed with Dulbecco's phosphate buffered saline (Dulbecco's Phosphate Buffered Saline, dPBS) and aspirated. Trypsin (3 mL per flask) was added and the flask was incubated for 2-3 minutes.
10mL of OptiMEM-10% fetal bovine-1% penicillin-streptomycin (hereinafter "OptiMEM medium") was added to the flask and cells and transferred to a 50mL conical tube. Cell counts (25 ul of cells in Effendor vials+25 ul of trypan blue stain) were performed and cell densities were adjusted to 15,000 cells/45 ul/well (3.33X10A 5/mL) in OptiMEM medium.
45 microliter of cell suspension (15,000 cells) was aliquoted into each well of a white 384-well plate. Plates were incubated for 10 minutes at room temperature before being placed in 37 ℃ plus 5% CO 2 The incubator was left overnight.
Day 2-compound treatment. Exemplary compounds were prepared at an initial concentration of 1mM and serial dilutions of 1:3 for 11 point CRC, prepared and stored in a refrigerator. The master compound plates were thawed overnight at room temperature. DMSO (20 μl) was added to column 24 of the master compound plate as a negative control, and 20 μl of 300 μΜ compound 4 was added to column 23 as a positive control.
The intermediate compound plates contained 4% DMSO in optmem medium. DMSO was added to the warmed optmem medium to obtain a 4% DMSO solution (approximately 50 mL/plate). 100 microliters of OptiMEM-4% DMSO was aliquoted into each well of a 384 well deep well microplate.
Spin-settling the main compound plate and the intermediate compound plate.
Transfer 1. Mu.l of compound in the master compound plate to the intermediate plate (1:100 dilution). The diluted mixtures were mixed and 5. Mu.L was transferred to assay plates (1:10 dilution) to give a final starting concentration of 1. Mu.M. The treated assay plate was incubated at 37℃with +5% CO 2 Incubate for 24 hours. The master compound plate was sealed and stored at room temperature for a second round within one week.
Day 3-HiBit assay. 5 microliters of Alamar Blue was added to each well of a black 384 well plate. Plates were incubated in an incubator (37 ℃ C.+5% CO 2 ) For 2 hours and for one hour at room temperature. The fluorescence of each plate was read on a plate reader for Alamar Blue viability assay.
A set of white assay plates was warmed to room temperature (45 minutes).
One Glo luciferase mixture was prepared. Media from a white 384 well assay plate was aspirated. mu.L of One Glo luciferase mix was added to each well of the assay plate. Plates were incubated on a bench (room temperature) for 45 minutes, including shaking at 700rpm for 10 minutes. The fluorescence of each plate is read on a plate reader.
1:100DLR substrate and 1:100LgBiT protein dilutions were added to Promega Stop and Glo buffers and mixed immediately prior to addition to the assay plates. To each well 25 microliters of Stop and Glo mixture was added. The assay plate was incubated for at least 45 minutes, including shaking at 700rpm for 10 minutes. The fluorescence of each plate is read on a plate reader.
Analysis of LRRK2 HiBit screening assay. As mentioned above, the plates were run in triplicate and the assay was repeated twice (6 replicates total for the exemplary compounds). For each treatment, firefly luciferase was measured for cell number, cell viability (Alamar Blue), and nanolu was measured for LRRK2-HiBit quantification.
For each plate, LRRK2 HiBit and alamar blue signals were normalized to median% DMSO. Curve fitting was performed for each compound to perform replicates on three plates. The data for the exemplary compounds of table 1 below are shown in the endogenous WT DC50 (nM) and endogenous WT Dmax (%) columns in table 2 below.
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
**D Max The range is as follows: a is more than or equal to 70; b is more than or equal to 50 and less than 70; c < 50
N.d.: not measured
A novel bifunctional molecule is described comprising an LRRK2 recruiting moiety and an E3 ubiquitin ligase recruiting moiety. The bifunctional molecules of the present disclosure actively degrade LRRK2, resulting in strong inhibition of cell proliferation and induction of apoptosis. Protein degradation mediated by the bifunctional compounds of the present disclosure provides a promising strategy for targeting "non-patentable" pathological proteins by conventional methods.
The foregoing general field of applicability is given by way of example only and is not intended to limit the scope of the disclosure and the appended claims. Further objects and advantages associated with the compositions, methods and processes of the present disclosure will be understood by those of ordinary skill in the art in light of the claims, specification and examples of the present invention. For example, the various aspects and embodiments of the disclosure may be used in a variety of combinations, all of which are explicitly contemplated by this specification. Such additional aspects and embodiments are expressly included within the scope of the present disclosure. Publications and other materials used herein to illuminate the background of the disclosure and in particular cases to provide additional details respecting the practice, are incorporated by reference.
Thus, those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the disclosure described herein. Such equivalents are intended to be encompassed by the following claims. It should be understood that the detailed examples and embodiments described herein are given by way of example only for illustrative purposes and are in no way to be construed as limiting the present disclosure. Various modifications or variations in light of the invention will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and are to be considered within the scope of the appended claims. For example, the relative amounts of the ingredients may be varied to optimize a desired effect, additional ingredients may be added, and/or similar ingredients may be substituted for one or more of the ingredients. Further advantageous features and functions associated with the systems, methods and processes of the present disclosure will be apparent from the appended claims. Furthermore, those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the disclosure described herein. Such equivalents are intended to be encompassed by the following claims.

Claims (49)

1. A heterobifunctional compound having the chemical structure:
PTM―L―CLM,
or a pharmaceutically acceptable salt or solvate thereof,
wherein:
(a) The CLM is a small molecule E3 ubiquitin ligase binding moiety that binds cereblon E3 ubiquitin ligase and is represented by the following chemical structure:
wherein:
w is CH 2 、C=O、SO 2 Or NH;
each X is independently selected from the group consisting of absence, O and S
Z is absent, O or S;
g is H or unsubstituted or substituted straight or branched alkyl;
each Q 1 、Q 2 、Q 3 And Q 4 N, CH or CR independently;
a is H or unsubstituted or substituted straight or branched alkyl;
n is an integer from 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched alkyl, optionally substituted alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L; and is also provided with
R 'and R' are independently selected from the group consisting of a bond, H, and optionally substituted alkyl;
represents a single bond or a double bond; and is also provided with
Represents a bond that may be stereotactic ((R) or (S)) or non-stereotactic;
(b) The PTM is a small molecule leucine rich repeat kinase 2 (LRRK 2) targeting moiety that binds to LRRK2 or a mutant form thereof, represented by the following chemical structure:
Wherein:
R 1 is isopropyl, tert-butyl,Wherein->Is the point of attachment to the oxygen atom of the PTM;
R 2 is hydrogen, F, cl, OH, C1-C3 alkyl or C1-C3 fluoroalkyl;
X 6 and X 7 Each independently is CH or N;
X 1 、X 2 、X 3 、X 4 and X 5 Each independently is N or CH, when CH, optionally R 2 Substitution;
X 8 is CH or N;
is an optionally substituted 3-10 membered cycloalkyl, heterocycloalkyl, bicycloalkyl, spirocycloalkyl or spiroheterocycloalkyl containing 1-4 (e.g., 1, 2, 3 or 4) heteroatoms selected from N, O and S (e.g., optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents); and is also provided with
The PTM isRepresents a point of attachment to the L; and is also provided with
(c) The L is a chemical linker group that covalently couples the CLM to the PTM.
2. The compound of claim 1, wherein the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
3. The compound of claim 1, wherein the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
4. A heterobifunctional compound having the chemical structure:
PTM―L―CLM,
or a pharmaceutically acceptable salt or solvate thereof,
wherein:
(a) The CLM is a small molecule E3 ubiquitin ligase binding moiety that binds cereblon E3 ubiquitin ligase and is represented by the following chemical structure:
Wherein:
w is CH 2 、C=O、SO 2 Or NH;
each X is independently selected from the group consisting of absence, O and S
Z is absent, O or S;
g is H or unsubstituted or substituted straight or branched alkyl;
each Q 1 、Q 2 、Q 3 And Q 4 N, CH or CR independently;
a is H or unsubstituted or substituted straight or branched alkyl;
n is an integer from 1 to 10 (e.g., 1-4, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10);
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched alkyl, optionally substituted alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L; and is also provided with
R 'and R' are independently selected from the group consisting of a bond, H, and optionally substituted alkyl;
represents a single bond or a double bond; and is also provided with
Represents a bond that may be stereotactic ((R) or (S)) or non-stereotactic;
(b) The PTM is a small molecule leucine rich repeat kinase 2 (LRRK 2) targeting moiety that binds to LRRK2 or a mutant form thereof, represented by the following chemical structure:
wherein:
R 1 is isopropyl, tert-butyl,Wherein->Is the point of attachment to the oxygen atom of the PTM;
R 2 is hydrogen, F, cl, OH, C1-C3 alkyl or C1-C3 fluoroalkyl;
X 6 and X 7 Each independently is CH or N;
X 1 、X 2 、X 3 、X 4 and X 5 Each independently is N or CH, when CH, optionally R 2 Substitution;
X 8 is CH or N;
is an optionally substituted 3-10 membered cycloalkyl, heterocycloalkyl, bicycloalkyl, spirocycloalkyl or spiroheterocycloalkyl containing 1-4 (e.g., 1, 2, 3 or 4) heteroatoms selected from N, O and S (e.g., optionally substituted with one or more (e.g., 1, 2, 3 or 4) substituents); and is also provided with
The PTM isRepresents a point of attachment to the L; and is also provided with
(c) The L is a bond or a chemical linker group that covalently couples the CLM to the PTM.
5. The compound according to claim 4, wherein the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
6. The compound according to claim 4, wherein the PTM is represented by:
wherein said PTMRepresents the point of connection to the L.
7. The compound of any one of claims 1-7, wherein the CLM is represented by the following chemical structure:
8. the compound of any one of claims 1-7, wherein the compound is represented by a chemical structure selected from the group consisting of:
wherein:
X 2 c, CH or N;
Z 1 is a bond, carbon shared with a cyclic group of L, or nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
R is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
9. The compound of any one of claims 1-7, wherein the compound is represented by a chemical structure selected from the group consisting of:
wherein:
X 2 c, CH or N;
Z 1 is a bond, carbon shared with a cyclic group of L, or nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
10. The compound of claim 10, wherein the compound is represented by the following chemical structure:
11. the compound of claim 10, wherein the compound is represented by the following chemical structure:
12. the compound of any one of claims 1-11, wherein each R is selected from H, O, OH, NH, NH 2 、-Cl、-F、CN、CF 3 Optionally substituted straight-chain or branched C 1-3 Alkyl, optionally substituted straight or branched C 1-3 An alkoxy group.
13. The compound of any one of claims 1-11, wherein each R is selected from H, O, OH, NH, NH 2 、-Cl、-F、-CN、CF 3 Methyl, methoxy and ethoxy.
14. The compound of any one of claims 1-13, wherein:
(a)is->
Wherein:
R 3 is H or methyl;
R 3a is H, halogen or methyl;
R 4 is H or methyl;
R 5 is H or methyl;
representing a point of connection to the PTM; and is also provided with
Represents the point of attachment of said L and when +.>In the absence of said-> Through atoms of N or CH, R of said cyclic group 3 、R 4 Or R is 5 Is connected to the L;
(b)R 2 is H or F; or (b)
(c) A combination thereof.
15. The compound of any one of claims 1-14, whereinSelected from the group consisting of: />
/>
Wherein said heterocycloalkyl is attached to said L or said PTM through an atom of said cyclic group or a substituent thereof.
16. The compound of any one of claims 1-13, wherein the PTM is represented by the following chemical structure:
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein the method comprises the steps ofRepresents the attachment site of the L.
17. The compound of any one of claims 1-16, wherein L comprises a chemical structural unit represented by the formula:
-(A L ) q -,
wherein:
-(A L ) q -is a group attached to the CLM and the PTM;
q is an integer greater than or equal to 1;
each A L Independently selected from CR L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 C.ident.C, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 0-6R L1 And/or R L2 Aryl substituted with groups and optionally with 1-6R L1 And/or R L2 A group-substituted heteroaryl group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
18. The compound of any one of claims 1-16, wherein the L comprises optionally substituted C 1 -C 50 Alkyl (e.g., C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 7 、C 8 、C 9 、C 10 、C 11 、C 12 、C 13 、C 14 、C 15 、C 16 、C 17 、C 18 、C 19 、C 20 、C 21 、C 22 、C 23 、C 24 、C 25 、C 26 、C 27 、C 28 、C 29 、C 30 、C 31 、C 32 、C 33 、C 34 、C 35 、C 36 、C 37 、C 38 、C 39 、C 40 、C 41 、C 42 、C 43 、C 44 、C 45 、C 46 、C 47 、C 48 、C 49 Or C 50 Alkyl), wherein:
Each carbon is optionally replaced by: CR (computed radiography) L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 C.ident.C, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 1-6R L1 And/or R L2 Aryl substituted by radicals or optionally substituted by 1-6R L1 And/or R L2 A group-substituted heteroaryl group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
19. The compound of any one of claims 1-18, wherein L is selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
the chemical linker group is optionally substituted with halogen;
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Each m, n, o, p, q and r of the L is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20.
20. The compound of any one of claims 1-19, wherein at least one of:
(a) The CLM is represented by:
/>
wherein: said ULMRepresents a point of attachment to the L; and N is a nitrogen atom shared with the L;
(b) The PTM is represented by:
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein the PTM is covalently linked to the L through an atom of the heterocycloalkyl a or a substituent thereof;
(c) The L is a linker group (L) selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Representing a point of attachment to said CLM or said PTM; or (b)
(d) A combination thereof.
21. The compound of claim 1, wherein at least one of:
the PTM is PTM selected from the group consisting of compounds 52-288;
the CLM is a CLM selected from compounds 52-288; and is also provided with
The L is L selected from compounds 52-288.
22. The compound of any one of claims 1-15, wherein one or more of:
g is H or unsubstituted or substituted straight-chain or branched C 1-6 An alkyl group;
a is H or unsubstituted or substituted straight-chain or branched C 1-6 An alkyl group;
r is a bond, H, O, -CONR 'R', -C (=O) R ', -OR', -NR 'R', unsubstituted OR substituted straight OR branched C 1-6 Alkyl, optionally substituted C 1-6 Alkoxy, -Cl, -F, -Br, -CF 3 or-CN, wherein one R is covalently bonded to said L;
r 'and R' are independently selected from the group consisting of bond, H and optionally substituted C 1-6 An alkyl group; and is also provided with
Is an optionally substituted 3-10 membered cycloalkyl, an optionally substituted 3-10 membered heterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered bicycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, an optionally substituted 3-10 membered spirocycloalkyl, or an optionally substituted 3-10 membered spiroheterocycloalkyl containing 1 to 4 (e.g., 1, 2, 3, or 4) heteroatoms, wherein said heteroatoms are independently selected from N, O and S.
23. The compound of any one of claims 1, 4, and 22, wherein the PTM has the chemical structure:
24. The compound of any one of claims 1, 4, and 23, wherein the compound has the following chemical structure:
wherein:
X 2 c, CH or N;
Z 1 is a bond, carbon shared with a cyclic group of L, or nitrogen shared with a cyclic group of L;
n is an integer from 0 to 3 (e.g., 0, 1, 2, or 3);
r is a bond, H, O, OH, N, NH, NH 2 Cl, -F, methyl, methoxy or ethoxy; and is also provided with
R 2 Is H, cl, F, OH, C C1-C3 alkyl or C1-C3 fluoroalkyl.
25. The compound of claim 24, wherein the compound is represented by the following chemical structure:
/>
26. the compound of claim 24, wherein the compound is represented by the following chemical structure:
27. the compound of any one of claims 23-26, wherein each R is selected from H, O, OH, NH, NH 2 、-Cl、-F、CN、CF 3 Optionally substituted straight-chain or branched C 1-3 Alkyl, optionally substituted straight or branched C 1-3 An alkoxy group.
28. The compound of any one of claims 23-26, wherein each R is selected from H, O, OH, NH, NH 2 、-Cl、-F、-CN、CF 3 Methyl, methoxy and ethoxy.
29. The compound of any one of claims 23-28, wherein:
(a)is->/>
Wherein:
R 3 is H or methyl;
R 3a is H, halogen or methyl;
R 4 Is H or methyl;
R 5 is H or methyl;
representing a point of connection to the PTM; and is also provided with
Representing and said LAnd when->In the absence of said-> Through atoms of N or CH, R of said cyclic group 3 、R 4 Or R is 5 Is connected to the L;
(b)R 2 is H or F; or (b)
(c) A combination thereof.
30. The compound of any one of claims 23-29, whereinSelected from the group consisting of: />
/>
Wherein said heterocycloalkyl is attached to said L or said PTM through an atom of said cyclic group or a substituent thereof.
31. The compound of any one of claims 1-15 and 22-30, wherein the CLM has a chemical structure represented by:
32. the compound of any one of claims 1-13 and 22-31, wherein:
(a)is->
Wherein:
R 3 is H or methyl;
R 3a is H, halogen or methyl;
R 4 is H or methyl;
R 5 is H or methyl;
representing a point of connection to the PTM; and is also provided with
Represents the point of attachment to said L and when +.>In the absence of said-> Through atoms of N or CH, R of said cyclic group 3 Or R is 4 Is connected to the L;
(b)R 2 is H or F; or (b)
(c) A combination thereof.
33. The compound of claim 32, whereinSelected from the group consisting of:
wherein said heterocycloalkyl is attached to said L or said PTM through an atom of said cyclic group or a substituent thereof.
34. The compound of any one of claims 1-13 and 22, wherein the PTM is represented by the following chemical structure:
/>
/>
wherein->Represents the attachment site of the L.
35. The compound of any one of claims 23-34, wherein L comprises a chemical structural unit represented by the formula:
-(A L ) q -,
wherein:
-(A L ) q -is a group attached to the CLM and the PTM;
q is an integer greater than or equal to 1;
each A L Independently selected from CR L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 C.ident.C, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 0-6R L1 And/or R L2 Aryl substituted with groups and optionally with 1-6R L1 And/or R L2 A group-substituted heteroaryl group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
36. The compound of any one of claims 23-34, wherein the L comprises optionally substituted C 1 -C 50 Alkyl (e.g., C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 7 、C 8 、C 9 、C 10 、C 11 、C 12 、C 13 、C 14 、C 15 、C 16 、C 17 、C 18 、C 19 、C 20 、C 21 、C 22 、C 23 、C 24 、C 25 、C 26 、C 27 、C 28 、C 29 、C 30 、C 31 、C 32 、C 33 、C 34 、C 35 、C 36 、C 37 、C 38 、C 39 、C 40 、C 41 、C 42 、C 43 、C 44 、C 45 、C 46 、C 47 、C 48 、C 49 Or C 50 Alkyl), wherein:
each carbon is optionally replaced by: CR (computed radiography) L1 R L2 、O、S、SO、SO 2 、NR L3 、SO 2 NR L3 、SONR L3 、CONR L3 、NR L3 CONR L4 、NR L3 SO 2 NR L4 、CO、CR L1 =CR L2 C.ident.C, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Cycloalkyl, optionally substituted with 1-9R L1 And/or R L2 Group-substituted C 5-13 Spirocycloalkyl, optionally substituted with 1-6R L1 And/or R L2 Group-substituted C 3-11 Heterocyclyl, optionally substituted with 1-8R L1 And/or R L2 Group-substituted C 5-13 Spiroheterocyclyl, optionally substituted with 1-6R L1 And/or R L2 Aryl substituted by radicals or optionally substituted by 1-6R L1 And/or R L2 A group-substituted heteroaryl group; and is also provided with
R L1 、R L2 、R L3 、R L4 And R is L5 Each independently is H, halogen, C 1-8 Alkyl, OC 1-8 Alkyl, SC 1-8 Alkyl, NHC 1-8 Alkyl, N (C) 1-8 Alkyl group 2 、C 3-11 Cycloalkyl, aryl, heteroaryl, C 3-11 Heterocyclyl, OC 3-8 Cycloalkyl, SC 3-8 Cycloalkyl, NHC 3-8 Cycloalkyl, N (C) 3-8 Cycloalkyl radicals) 2 、N(C 3-8 Cycloalkyl) (C) 1-8 Alkyl), OH, NH 2 、SH、SO 2 C 1-8 Alkyl, P (O) (OC 1-8 Alkyl) (C) 1-8 Alkyl), P (O) (OC 1-8 Alkyl group 2 、CC-C 1-8 Alkyl, CCH, ch=ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =ch (C 1-8 Alkyl group), C (C) 1-8 Alkyl) =c (C 1-8 Alkyl group 2 、Si(OH) 3 、Si(C 1-8 Alkyl group 3 、Si(OH)(C 1-8 Alkyl group 2 、COC 1-8 Alkyl, CO 2 H、CN、CF 3 、CHF 2 、CH 2 F、NO 2 、SF 5 、SO 2 NHC 1-8 Alkyl, SO 2 N(C 1-8 Alkyl group 2 、SONHC 1-8 Alkyl, SON (C) 1-8 Alkyl group 2 、CONHC 1-8 Alkyl, CON (C) 1-8 Alkyl group 2 、N(C 1-8 Alkyl) CONH (C 1-8 Alkyl), N (C) 1-8 Alkyl) CON (C 1-8 Alkyl group 2 、NHCONH(C 1-8 Alkyl), NHCON (C) 1-8 Alkyl group 2 、NHCONH 2 、N(C 1-8 Alkyl) SO 2 NH(C 1-8 Alkyl), N (C) 1-8 Alkyl) SO 2 N(C 1-8 Alkyl group 2 、NH SO 2 NH(C 1-8 Alkyl, NH SO 2 N(C 1-8 Alkyl group 2 Or NH SO 2 NH 2
37. The compound of any one of claims 1-13 and 23-36, wherein L is selected from the group consisting of:
/>
/>
wherein:
the chemical linker group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen and methyl (preferably independently selected from halogen);
c is a carbon atom covalently linked or shared with said CLM or said PTM;
n is a nitrogen atom covalently linked or shared with said CLM or said PTM; and is also provided with
Each m, n, and o of the L is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.
38. The compound of any one of claims 1-37, wherein at least one of:
(a) The CLM is represented by:
/>
/>
/>
wherein:
said ULMRepresents a point of attachment to the L; c is a carbon atom shared with the L; and N is a nitrogen atom shared with the L;
(b) The PTM is represented by:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein the PTM is covalently linked to the L through an atom of the heterocycloalkyl a or a substituent thereof;
(c) The L is a linker group (L) selected from the group consisting of:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
wherein:
n is a nitrogen atom covalently linked or shared with said CLM or said PTM;
c is a carbon atom covalently linked or shared with said CLM or said PTM; and is also provided withRepresenting a point of attachment to said CLM or said PTM; or (b)
(d) A combination thereof.
39. The compound of claim 1, wherein at least one of:
the PTM is a PTM selected from the compounds of table 1;
the CLM is a CLM selected from the compounds of table 1; and is also provided with
The L is L selected from the compounds of Table 1.
40. The compound of claim 1, wherein the compound is represented by the following chemical structure:
wherein:
n is 0 or 1;
r is H, OH, -Cl, -F or Br;
Z 1 is nitrogen or carbon shared with the cyclic group of L;
X 2 is C or N;
X 4 is CH or N (preferably N);
R 1 is that(preferably->) Wherein->Is the point of attachment to the oxygen of the PTM;
R 2 Is H, -Cl or-F;
the method comprises the following steps: />
(preferably->
) Wherein:
R 3 is H, methyl or ethyl;
R 4 is H,Methyl or ethyl;
representing said->A connection point with the PTM; and is also provided with
Represents the junction of said PTM with said L and when +.>In the absence of said->May be attached to the L through an atom of the cyclic group (e.g., carbon or nitrogen);
l is represented by the following chemical structure:
/>
wherein:
m and n are integers independently selected from 0, 1, 2 or 3 (preferably 1); and is also provided with
The L is optionally substituted with 0, 1, 2 or 3 (preferably 0 or 1) groups selected from: -Cl, -F and C 1-3 Alkyl (e.g., methyl or ethyl).
41. The compound of claim 40, wherein X 2 Is C and X 4 Is N.
42. The compound of claim 1, wherein the compound is selected from the group consisting of compounds 52-384 of table 1:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
or an enantiomer thereof.
43. A compound selected from the group consisting of:
or an enantiomer thereof.
44. A composition comprising an effective amount of the bifunctional compound of any one of claims 1-43 and a pharmaceutically acceptable carrier.
45. The composition of claim 44, wherein the composition further comprises an additional bioactive agent.
46. The composition of claim 45, wherein the additional bioactive agent is an anti-inflammatory agent, a chemotherapeutic agent, or an immunomodulatory agent.
47. A composition comprising a pharmaceutically acceptable carrier and an effective amount of at least one compound of any one of claims 1-43 for use in treating a disease, disorder, or symptom of a causal relationship with LRRK2 in a subject, wherein the composition is effective to treat or ameliorate the disease, disorder, or at least one symptom of the disease or disorder.
48. The composition of claim 47, wherein the disease or disorder is idiopathic Parkinsonism (PD), LRRK2 mutation related PD, primary tauopathy, lewy body dementia, crohn's disease, leprosy, neuroinflammation, progressive supranuclear palsy, pick's disease, FTDtau, TDP-43 frontotemporal dementia, TDP-43ALS, c9orf ALS, huntington's disease, spinocerebellar ataxia (SCAs) 1, 2, 3, 6, 7 and 17, dentate nuclear pallidoluffing (DRPLA) or Kenydi's disease.
49. A method of treating or preventing a disease, disorder, or condition associated with LRRK2, the method comprising providing to a patient in need thereof an effective amount of a compound as described herein or a composition comprising the compound and administering to the patient an effective amount of a compound as described herein or a composition comprising the compound, wherein the compound or composition is effective to treat or ameliorate the disease, disorder, or at least one symptom of the disease or disorder.
CN202280035511.XA 2021-03-19 2022-03-19 Indazole-based compounds and related methods of use Pending CN117321046A (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US63/163,328 2021-03-19
US63/228,731 2021-08-03
US63/243,014 2021-09-10
US202163245411P 2021-09-17 2021-09-17
US63/245,411 2021-09-17
PCT/US2022/021049 WO2022198112A1 (en) 2021-03-19 2022-03-19 Indazole based compounds and associated methods of use

Publications (1)

Publication Number Publication Date
CN117321046A true CN117321046A (en) 2023-12-29

Family

ID=89288873

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280035511.XA Pending CN117321046A (en) 2021-03-19 2022-03-19 Indazole-based compounds and related methods of use

Country Status (1)

Country Link
CN (1) CN117321046A (en)

Similar Documents

Publication Publication Date Title
JP7041198B2 (en) 5-Chloro-2-difluoromethoxyphenylpyrazolopyrimidine compound which is a JAK inhibitor
TWI765908B (en) Benzimidazole compounds as kinase inhibitors, and preparation methods and applications thereof
DK2545045T3 (en) PIPERIDINE-4-YL-azetidine derivatives AS JAK1 INHIBITORS
CN112368283B (en) Bicyclic derivative-containing inhibitor, preparation method and application thereof
KR20190012167A (en) Isoquinolin-3-ylcarboxamide, its preparation and uses
KR20190014505A (en) Isoquinolin-3-ylcarboxamide, its preparation and uses
WO2006093247A1 (en) NOVEL AMINOPYRIDINE COMPOUND WITH Syk INHIBITORY ACTIVITY
KR20100099742A (en) Bicyclic derivatives for use in the treatment of androgen receptor associated conditions
BR112013014854B1 (en) n- (1h-indazol-4-yl) imidazo [1,2-a] pyridine-3-carboxamide compounds substituted as type iii receptor tyrosine kinase inhibitors, their production process and pharmaceutical composition
EP3999182A1 (en) Tau-protein targeting compounds and associated methods of use
US20230097358A1 (en) Indazole based compounds and associated methods of use
JP6472803B2 (en) Triazolopyridine compounds, compositions thereof and methods of use
KR20230004511A (en) Indazole-based compounds and related methods of use
JP2023518831A (en) Selective modulators of mutant LRRK2 proteolysis and related methods of use
WO2006112828A1 (en) Azaindole derivatives as inhibitors of p38 kinase
US20190337946A1 (en) Triazolopyridine compounds and methods of use thereof
CN117321046A (en) Indazole-based compounds and related methods of use
AU2017324281A1 (en) 8-(azetidin-1-yl)-[1,2,4]triazolo[1,5-A]pyridinyl compounds, compositions and methods of use thereof
AU2019257385B2 (en) Piperidin-4-yl azetidine derivatives as jak1 inhibitors
CN115843296B (en) CDK9 inhibitors and uses thereof
WO2023115150A1 (en) Bifunctional arylsulphonamide compounds

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination