NZ625745B2 - FGFR antibody drug conjugates (ADCs) and the use thereof - Google Patents

FGFR antibody drug conjugates (ADCs) and the use thereof Download PDF

Info

Publication number
NZ625745B2
NZ625745B2 NZ625745A NZ62574512A NZ625745B2 NZ 625745 B2 NZ625745 B2 NZ 625745B2 NZ 625745 A NZ625745 A NZ 625745A NZ 62574512 A NZ62574512 A NZ 62574512A NZ 625745 B2 NZ625745 B2 NZ 625745B2
Authority
NZ
New Zealand
Prior art keywords
group
marks
hydrogen
formula
linkage site
Prior art date
Application number
NZ625745A
Other versions
NZ625745A (en
Inventor
Sandra Bruder
Sven Golfier
Simone Greven
Stefanie Hammer
Axel Harrenga
Charlotte Christine Kopitz
Hans Georg Lerchen
Christoph Mahlert
Carl Friedrich Nising
Joachim Schuhmacher
Original Assignee
Seattle Genetics Inc
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 Seattle Genetics Inc filed Critical Seattle Genetics Inc
Priority claimed from PCT/EP2012/075277 external-priority patent/WO2013087716A2/en
Publication of NZ625745A publication Critical patent/NZ625745A/en
Publication of NZ625745B2 publication Critical patent/NZ625745B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K11/00Depsipeptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof

Abstract

Disclosed are antibody-linked auristatin conjugates of the general formula (Ia) which comprise a novel linker, wherein AK is a binder which is an antibody or an antigen-binding antibody fragment that binds to FGFR2, and wherein the linker comprises the group L2 which is a linear C2-C10alkylene-##4 group, or a polyethylene linking group of the formula ##3-CH2-CH2-[O-CH2-CH2-]p-##4; p is a number from 2 to 6; ##3 marks the linkage site on the side with AK; ##4 marks the bond to the nitrogen atom of the auristatin derivative, and wherein the C2-C10alkylene group is optionally substituted by 1 to 4 substituents selected independently of one another from the group consisting of methyl, hydroxyl, and benzyl, or wherein two carbon atoms of the C2-C10alkylene group in 1,2-, 1,3- or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, are optionally bridged to form a cycloalkyl ring or a phenyl ring; for the treatment and/or prophylaxis of hyperproliferative and/or angiogenic diseases. In a preferred embodiment L2 is an unsubstituted pentylene chain linked to AK through a carbonyl group. roup, or a polyethylene linking group of the formula ##3-CH2-CH2-[O-CH2-CH2-]p-##4; p is a number from 2 to 6; ##3 marks the linkage site on the side with AK; ##4 marks the bond to the nitrogen atom of the auristatin derivative, and wherein the C2-C10alkylene group is optionally substituted by 1 to 4 substituents selected independently of one another from the group consisting of methyl, hydroxyl, and benzyl, or wherein two carbon atoms of the C2-C10alkylene group in 1,2-, 1,3- or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, are optionally bridged to form a cycloalkyl ring or a phenyl ring; for the treatment and/or prophylaxis of hyperproliferative and/or angiogenic diseases. In a preferred embodiment L2 is an unsubstituted pentylene chain linked to AK through a carbonyl group.

Description

FGFR antibody drug conjugates (ADCs) and the use thereof The present application s to new binder-drug ates (ADCs) of N,N-dialkylauristatins that are directed against the target fibroblast growth factor receptor 2 (FGFR2), to active metabolites of these ADCs, to processes for preparing these ADCs, to the use of these ADCs for treating and/or preventing illnesses, and also to the use of these ADCs for producing medicaments for treating and/or preventing illnesses, more ularly roliferative and/or angiogenic es such as, for example, cancer diseases. Such treatments may be sed as a monotherapy or else in combination with other medicaments or further therapeutic measures.
Cancer diseases are the consequence of uncontrolled cell growth in a wide variety of tissues. In many cases the new cells penetrate existing tissue (invasive growth), or they metastase into remote organs. Cancer diseases occur in a wide variety of organs, and the illnesses often ss in a tissue-specific manner. The designation “cancer disease” as a generic term therefore describes a large group of defined diseases of different organs, tissues and cell types.
Early-stage tumours may be able to be removed by al and radiotherapeutic measures.
Metastasized tumours can generally only be given palliative therapy by means of chemotherapeutic agents. The objective in that case is to achieve the optimum combination of ing quality of life and prolonging remaining lifetime.
The majority of the chemotherapeutic agents which are presently administered parenterally are often not target-directed at the tumour tissue or the tumour cells, but instead, as a result of their systemic administration, are buted non-specifically within the body, hence ing at locations at which exposure to the drug is undesirable, such as in healthy cells, tissues and organs, for example. This may lead to ed side-effects and even to serious effects of l toxicity, which then often greatly limit the therapeutically useful dose range of the drug, or necessitate complete cessation of medication.
The improved and selective availability of these chemotherapeutic agents in the tumour cell or the immediately surrounding , and the associated boost in effect, on the one hand, and minimization of toxic side-effects, on the other hand, have therefore been a focal point for a number of years in the development of new chemotherapeutic agents. Many attempts have been made to date to develop efficient methods of introducing the drug into the target cell. Optimizing the association between drug and intracellular target and minimizing the intercellular distribution of drug, to adjacent cells, for e, nevertheless continue to constitute a difficult problem.
Monoclonal antibodies, for example, are suitable for the target-directed sing of tumour tissue and tumour cells. The significance of such antibodies for the clinical treatment of cancer es has seen a erable general increase in recent years, based on the activity of such agents as trastuzumab (Herceptin®), mab (Rituxan®), cetuximab (Erbitux®) and bevacizumab (Avastin ®), which have since been approved for the therapy of individual, specific tumour es [see e.g. G. P. Adams and L. M. Weiner, Nat. Biotechnol. 23, 1147-1157 (2005)]. Consequently there has also been a marked increase in interest in so-called immunoconjugates such as, for example, the aforementioned ADCs, in which an internalizing antibody directed against a tumourassociated antigen is joined covalently via a linking unit ("linker") to a cytotoxic agent. Following introduction of the ADC into the tumour cell and subsequent cleavage of the conjugate, either the cytotoxic agent itself or another metabolite with cytotoxic activity, formed from the cytotoxic agent, is released within the tumour cell, where it is able to develop its effect directly and selectively. In this way it would be possible to keep the damage to normal tissue within significantly closer limits in comparison to a conventional chemotherapy of the cancer disease [see e.g. J. M. Lambert, Curr. Opin. Pharmacol. 5, 543-549 (2005); A. M. Wu and P. D. Senter, Nat.
Biotechnol. 23, 1137-1146 (2005); P. D. Senter, Curr. Opin. Chem. Biol. 13, 235-244 (2009); L. Ducry and B. Stump, Bioconjugate Chem. 21, 5-13 (2010)].
Instead of antibodies, it is also possible for binders from the small-molecule drug sphere to be used as binders which bind ively to a specific target location ("target"), such as to a or, for example [see e.g. E. Ruoslahti et al., Science 279, 377-380 (1998); D. Karkan et al., PLoS ONE 3 (6), e2469 (June 25, 2008)]. Also known are conjugates of cytotoxic drug and addressing ligand that exhibit a defined cleavage point between ligand and drug for the release of the drug. A "predetermined break point" of this kind may exist, for example, within a peptide chain which can be cleaved selectively at a particular site by a specific enzyme at the location of action [see e.g. R. A. Firestone and L. A. Telan, US Patent Application US 2002/0147138].
The activity of monoclonal antibodies is demonstrated in various types of cancer. Thus HERCEPTIN® and x® are used sfully in the treatment of HER2-positive breast cancer and EGFR-positive colorectal cancer, respectively.
The coupling of xic compounds to dies forms an extended possibility for additionally improving cancer therapy, since these conjugates allow tumour-specific toxophore lation and at the same time reduce the systemic toxicity. With respect to activity and nce, clinical studies with brentuximab vedotin in Hodgkin’s lymphoma and with trastuzumab-DM1 in breast cancer have yielded highly ing results, which support the development of new antibodies and new ADCs against other tumour antigens.
Antibody-based y proves to be very powerful in the treatment of various carcinomas, including solid tumours. Thus, for e Herceptin ® has been deployed sfully in the treatment of breast cancer, and Rituxan® is powerful in forms of carcinoma associated with the B cells. At the focal point of the pment of a sful antibody-based therapy is the isolation of antibodies against cell surface proteins which are expressed preferably on tumour cells.
The fibroblast growth factor receptors are tyrosine receptor kinases (RTK), of which four are known in mammals (FGFR1, FGFR2, FGFR3, FGFR4). As ligands, 22 human fibroblast growth factors (FGF) have been identified (Eswarakumar and Schlessinger, Cytokine & Growth Factor Reviews 2005, 16:139-149; Shimada et al., Proc. Natl. Acad. Sci. USA 2001, 98:6500-6505). The FGFRs consist of three extracellular immunoglobulin (Ig)-like domains, namely D1-D3, with domains 2 and 3 being necessary for ligand binding, and also of an individual transmembrane domain and a cytoplasmic domain, which contains the catalytic centre of the n tyrosine kinase (a schematic illustration is given in Figure 1).
Schematic illustration of the structure of FGFR2. Alpha (SEQ ID NO: 1) and beta (SEQ ID NO: 2) splice variants are compared with one another. The illustration shows the three Ig-like domains (D1, D2 and D3), the transmembrane domain (TM) and the intracellular kinase domain. The heparin binding site (HBS), the acidic box (AB), and the alternative IIIb/IIIc domains are likewise ed. The amino terminus is marked by N, the carboxy terminus by C.
The extracellular component additionally harbours the acidic box (AB) and the heparin binding site (HBS) (see Figure 1). An ant feature of the FGFR family of RTKs is that there are ent, alternatively d variants in existence. The full-length FGFR is identified as FGFR alpha (SEQ ID NO: 1), while the isoform, which is missing D1, is identified as FGFR beta (SEQ ID NO: 2) (Figure 1). An alternative splicing in domain 3 leads to two different variants, namely FGFR2 IIIb, which is encoded by the exons 7 and 8, and FGFR2 IIIc, which is encoded by the exons 7 and 9 (Figure 1). The latter splicing influences the ligand binding, leading to the specificity pattern. FGFR2 IIIc is expressed primarily by mesenchymal cells, and FGFR2 IIIb essentially by epithelial cells. FGF7 is also known as keratinocyte growth factor (KGF), and binds only to FGFR2 IIIb, which is therefore also called KGFR. When the FGFs bind to their receptors, there is subsequent zation and phosphorylation of the FGFRs, and a downstream signalling via the FRS-GRB2 docking n complex to the RAS-MAPK signal transduction cascade and the PI3K-AKT signal transduction cascade. The former signal transduction cascade is involved in cell growth and in cell entiation, while the latter is involved in the survival of the cells and in the ination of cell fate (Katoh und Katoh, Int. J. Oncol. 2006, 29:163-168).
For correct genesis during embryogenesis, an orchestrated signal transduction of all four receptors (FGFR1 to FGFR4) and of their splicing variants via the different FGFs is necessary (Ornitz et al., Genome Biol 2001, 2:3005). In the case of FGFR2, the absence of all FGFR2 variants leads to s in placental and limb bud formation, and therefore leads to fatality at the E10.5 stage. Specific knock-out of FGFR2 IIIb in the mouse leads likewise to fatality (at P0) in WO 87716 association with agenesis of the lungs, of the anterior lobe, of the thyroid gland, of the teeth and of the limbs, whereas disruption of the FGFR2 IIIc variant is survivable, with incidence of retarded ossification, tional dwarfism and synostosis of the base of the skull (Eswarakumar and Schlessinger, 2005). Activating mutations of FGFR2 in the germ track lead to severe malformations during embryogenesis, such as coronal osis and cranial synostosis in the case of Apert’s syndrome or in the case of Pfeiffer’s syndrome in humans (Robin et al., in Gene Reviews, NCBI elf Washington, edited by Pagon et al., 1993). In adults, FGFR2 signal transduction is involved in wound healing, in epithelial repair and in the protection of cells of the skin and mucous membrane (Braun et al., Phil. Trans. R. Soc. Lond. B 2004, 359:753-757) and in ration in the event of liver damage (Steiling et al., Oncogene 2003, 0-4388; Böhm, dissertation, Eidgenössische Technische hule Zürich, 2009). A role of FGFR2 signal transduction in the ion of EPDC (epicardial derived cells) into the heart following tion is therefore under discussion because during embryogenesis, FGF10-FGFR2 signal transduction is necessary for the migration of EPDC into the compact myocardium, a process which is necessary for the development of the heart Hernández et al., Development 2011:3331-3340; Winter and De Groot, Cell Mol. Life Sci. 2007, 64:692-703). All of these roles played by FGFR2 are regenerative in nature, and apparently of essential importance only under non-physiological conditions, as a result of a disruption in tissue homeostasis. Increased somatic signal transduction via FGFR2 is involved in various pathological conditions such as acne (Katoh, J. of Invest.
Dermatol. 2009, 129:1861-1867), psoriasis (Finch et al., Am. J. Pathol. 1997, 151:1619-1628; Xu et al., J. Invest. Dermatol. 2011:131:1521-1529) and cancer (see below).
A number of studies have been published which show a strong association of FGFR2 expression with an adverse outcome in cancer patients: The overexpression of FGFR2 and/or KGF is accompanied by expansive growth of stomach carcinoma and shorter survival of the patients (Matsunobu et al., Int. J. Cancer 2006, 28:307-314; wa et al., Oncol. Reports 2009, 21:875-880). The overexpression of FGFR2 was detected in % of all h carcinoma samples examined (Matsunobu et al., Int. J. Cancer 2006, 28:307-314; Toyokawa et al., Oncol. Reports 2009, 21:875-880). Adenocarcinoma (70% of all stomach carcinomas) is subdivided, additionally, into two different ogical types, namely stomach cancer of the intestinal type of and of the diffuse type. Interestingly, the first, less aggressive type is associated with an ted ErbB2 signal pathway, whereas, in the case of the latter, more aggressive type, aberrations occur in the FGFR2/PI3K signal pathway (Yamashita et al., Surg. Today 2011, 41:24-38). FGFR2 overexpression occurred in 53% of all samples of stomach cancer of the e kind (Yamashita et al., Surg. Today 2011, 38). Drawing together all of the data, HER2 expression and FGFR2 expression appear to occur in two different patient populations. To some extent possibly, the expression of FGFR2 is the result of a gene amplification, since amplifications of FGFR2 are found in approximately 7-10% of all primary stomach carcinomas (Kunii et al., Cancer Res. 2008, 68:232348). Moreover, FGFR2 expression has not only been found in ases, but was in fact even greater in metastases than in primary tumours (Yamashita et al., Surg. Today 2011, 41:24-38).
In the case of breast cancer, FGFR2 IIIb expression was found in 57% of tumour samples, but hardly at all in healthy tissue (Tamaru et al. 2004, 84:1460-1471). KGF (FGF7) occurred in 45% of random samples, generally together with FGFR2 IIIb. The co-expression of FGF7 and its single receptor FGFR2 IIIb was associated with a significantly reduced number of tic cells within the primary tumour by comparison with primary breast carcinomas, where neither FGF7 nor FGFR2 IIIb was expressed (Tamaru et al. 2004, 84:1460-1471). As in the case of stomach cancer, a gene amplification was found for breast cancer as well, and also in 4% of triply negative breast carcinomas (TNBC) (Turner et al., Oncogene 2010, 29:2013-2023). In breast cancer, a number of changes in individual nucleotides (Single Nucleotide Polymorphism, SNP) have been fied that are associated with an sed risk of breast cancer (Hunter et al., Nature Genetics 2007, 6:870-874). If the SNPs are localized within Intron 2, this leads to a transcriptional upregulation of FGFR2 (Katoh, Expert Reviews 2010, 10:1375-1379). Interestingly, FGFR1 is upregulated preferentially in the case of gen receptor (ER) positive breast carcinomas, while FGFR2 is upregulated in the case of ER-negative breast omas (Katoh, Expert Reviews 2010, 10:1375- 1379).
In the case of pancreatic carcinoma, the overexpression of FGFR2 IIIb and/or FGF7 is heavily correlated with venal invasion (Cho et al., Am. J. Pathol. 170:1964-1974), with co-expression of FGFR2 and FGF7 having been found in tumour cells, but ing even more frequently in the stroma cells which are adjacent to the tumour cells ata et al., Am. J. Pathol. 1998, 3- 222).
In the case of epithelial ovary cancer, 80% of cases showed upregulation of FGRF2 as compared with normal tissue, and in 70% there was FGF7 in the ascitic fluid (Steele et al., Oncogene :5878-5887).
The FGFR2 protein was found in all invasive al carcinomas , with strong expression at the invasive front of the tumours (Kawase et al., Int. J. Oncol. 2010, 36:331-340).
In the case of pulmonary adenocarcinoma, co-expression of FGF7 and FGFR2 occurred in 51.6% of cases, and is correlated with lower degrees of differentiation, higher rate of proliferation, lymph node metastasis, and shorter 5-year survival (Yamayoshi et al., J. Pathol. 2004, 204:110-118).
In the case of endometrial carcinoma, activating FGFR2 mutations occur in approximately 16% of cases (Pollock et al., Oncogene 2007, 26:7158-7162).
In the case of oesophageal cancer (OC), co-expression of FGF7 and FGFR2 in cancer cells was found in 26% of patients, and was ated with a trend towards a shorter survival time (Yoshino et al., Int. J. Oncol. 2007, 31:721-728).
In the case of liver cell carcinoma, the expression of FGFR2 was lated 4.7 times more strongly in poorly differentiated s. This expression is ated with the incidence of portal vein invasion and lower disease-free survival times (Harimoto et al., Oncology 2010, 78:361-368).
A number of publications, with in vitro and in vivo experimental data, show a causal connection between altered FGFR2 signal transduction and tumour growth.
Knock-down or inhibition of FGFR2 in cells of h cancer (Takeda et al., Clin. Cancer Res. 2007; 13:3051-3057; Kunii et al., Cancer Res. 2008; 68:2340-2348), breast cancer (Turner et al., Oncogene 2010, 29:2013-2023), n cancer (Cole et al., Cancer Biol. Ther. 2010, 10:495-504) and squamous carcinoma of the head and neck all et al., Clin. Cancer Res. 2011, 17:5016- 5025) led to reduced proliferation or increased apoptosis of the tumour cells. In tumour xenotransplants as well, knock-down of FGFR2 and inhibition of FGFR2 in tumour cell lines which press FGFR2 was observed to cause growth inhibition in stomach cancer cell lines (Takeda et al., Clin. Cancer Res. 2007; 13:3051-3057) and ovarian cancer cell lines (Cole et al., Cancer Biol. Ther. 2010, 10:495-504). er, FGF7, which exclusively activates FGFR2, increases the proliferation of stomach cancer cell lines (Shin et al., J. Cancer Res. Clin. Oncol. 2002, 128:596–602), breast cancer cell lines (Zhang et al., ncer Res. 1998, 18:2541-2546) and ovarian cancer cell lines (Cole et al., Cancer Biol. Ther. 2010, 10:495-504) in vitro and in vivo . Furthermore, knock-down of FGFR2 in endometrial c ancer cell lines which contain FGFR2 with activating mutations likewise led to the standstill of the cell cycle and to the induction of cell death (Byron et al., Cancer Res. 2008, 68:6902-6907).
FGFR2 signal transduction promotes the migration and invasion of stomach cancer cell lines (Shin et al., J. Cancer Res. Clin. Oncol. 2002, 128:596–602), breast cancer cell lines (Zhang et al., Anticancer Res. 1998, 1-2546) and pancreatic cancer cell lines in vitro (Nomura et al., Br. J.
Cancer 2008, 99:305-313; Niu et al., J. Biol. Chem. 2007, 282:6601-6011).
In the case of oesophageal cancer, FGFR2 is the most highly upregulated gene in tumourassociated fibroblasts. Isolated tumour-associated fibroblasts released a e factor which promotes the proliferation of oesophageal cancer cells (Zhang et al., hum. Cancer Biol. 2009, :4017-4022), thereby demonstrating that FGFR2 expressed by stroma cells is also able to promote tumour progression.
There are only a few reports on anti-FGFR2 antibodies. Fortin et al. (J. Neurosci. 2005, 25: 7470- 7479) describe a blocking anti-FGFR2 antibody. Wei et al. (Hybridoma 2006, 25: 115-124) have shown antibodies with exclusive specificity for FGFR2 IIIb, which inhibit the KGF-induced cell proliferation. WO2007/144893 describes inhibiting antibodies which bind FGFR2 and FGFR3. In WO2010/054265 and in Zhao et al. (Clin. Cancer Res. 2010,16:5750-5758), antibodies which inhibit FGF binding are described, including for example GAL-FR21 and GAL-FR22. Bai et al.
(Cancer Res. 2010, 0-7639) describe antibodies having specificity for FGFR2 IIIb. R&D Systems market GFR2 antibodies which have an ty-neutralizing effect in the cturer’s assays. WO2005/066211 describes dies which are directed against various cell-surface FGFRs, including FGFR2. WO2009/100105 describes isoform-specific anti-FGFR2 antibodies which can be linked covalently to effector molecules. WO2007/134210 describes methods for treating colorectal cancer using anti-FGFR2 antibodies or immunoconjugates.
WO2007/144893 describes FGFR2 antibodies with binding ty for further FGFRs, which block the ligand-dependent and the constitutive ligand-independent FGFR2 receptor tion.
Auristatin E (AE) and monomethylauristatin E (MMAE) are synthetic analogues of the dolastatins, a specific group of linear pseudopeptides which were originally isolated from marine sources and which have in some cases very potent cytotoxic activity with respect to tumour cells [for a review see e.g. G. R. Pettit, Prog. Chem. Org. Nat. Prod. 70, 1-79 (1997); G. R. Pettit et al., Anti-Cancer Drug Design 10, 529-544 (1995); G. R. Pettit et al., Anti-Cancer Drug Design 13, 243-277 (1998)].
H3C CH H C 3 CH3 OH O H H N R N N N N O O CH CH O CH O O 3 3 3 CH3 3 CH 3 CH Auristatin E (AE): R = CH3 Monomethylauristatin E (MMAE): R = H MMAE, however, possesses the disadvantage of a comparatively high systemic toxicity. For ing tumour selectivity, MMAE is used more particularly in conjunction with enzymatically cleavable -citrulline linkers in the ADC setting for more ed tumour therapy [-A2; S. O. Doronina et al., Bioconjugate Chem. 17, 114-124 (2006)]. Following proteolytic ge, MMAE is released preferably intracellularly from corresponding ADCs.
Monomethylauristatin F (MMAF) is an auristatin derivative having a C-terminal alanine unit which exhibits only moderate antiproliferative activity in comparison to MMAE. This fact is very probably attributable to the free carboxyl group, whose ty and charge adversely affect the capacity of this compound to access cells. In this connection, the methyl ester of MMAF (MMAF-OMe) has been described, as a l-charged prodrug tive with cell access capability, which, in comparison to MMAF, has an in vitro cytotoxicity for various carcinoma cell lines that is increased by a number of orders of magnitude [S. O. Doronina et al., Bioconjugate Chem. 17, 114-124 (2006)]. It can be d that this effect is brought about by MMAF itself, which, following uptake of the prodrug into the cells, is rapidly released by intracellular ester hydrolysis.
H C CH H3C CH 3 3 O O 3 H H N N N O HN N O O CH O CH O O 3 3 CH 3 CH 3 CH 3 Monomethylauristatin F (MMAF): R = H Monomethylauristatin F methyl ester (MMAF-OMe): R = CH3 However, drug compounds based on simple ester tives are generally subject to the risk of al instability on account of non-specific ester ysis, independent of the intended site of action, by means, for example, of esterases that are present in the blood plasma; this non-specific hydrolysis may significantly restrict the usefulness of such compounds in therapy.
Monomethylauristatin F (MMAF) and also various ester derivatives and amide derivatives thereof have been disclosed in -A2. Further auristatin analogues with a C-terminal, amidically substituted phenylalanine unit are described in WO 01/18032-A2. WO 02/088172-A2 and WO 08603-A1 claim MMAF analogues which relate to side-chain modifications of the phenylalanine, while -A2 claims those in which the carboxyl group of the alanine has been modified. Auristatin conjugates linked via the C-terminus have been recently described in WO 17531-A1 [see also S. O. Doronina et al., Bioconjugate Chem. 19, 1960-1963 (2008)].
The problem addressed with the present invention was that of ing new binder-drug conjugates (ADCs) which, through combination of new N,N-dialkylauristatin tives with innovative, suitable linkers and binder, exhibit a very attractive activity e, such as, for example, in terms of their specific tumour effect and/or the reduced potential of the metabolites formed intracellularly to be a substrate with respect to transporter proteins, and which are therefore suitable for the treatment and/or prophylaxis of hyperproliferative and/or angiogenic diseases, such as cancer diseases, for example. - 9 – the group -B- L2 -§§ is a linker, where WO 87716 where two carbon atoms of the alkanediyl chain in 1,2-, 1,3- or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, may be bridged to form a (C3-C6)-cycloalkyl ring or a phenyl ring, D is a group of the formula O #3 T1 #3 R5 N or #3 R3 R4 O , R26 T2 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen or methyl, R2 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, oxyethyl, 4-hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, 1-phenylethyl, diphenylmethyl, dazolylmethyl or 1H-indol ylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or ic, optionally substituted heterocycle of the formula O or O O O N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy or benzyloxy, R3 is hydrogen or methyl, R4 is isopropyl, isobutyl, sec-butyl, utyl, phenyl, , 1-hydroxyethyl, 4-hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, 1-phenylethyl, diphenylmethyl, 1H-imidazolylmethyl or 1H-indol ylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the e site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9, -C(=O)-NH-NH-R10 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen or methyl, R9 is hydrogen, methyl, ethyl, n-propyl or benzyl, R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to 7-membered cycle, R10 is benzoyl, R11 is benzyl, which may be substituted in the phenyl group by methoxycarbonyl or yl, R5 is hydrogen, methyl or a group of the formula #9 O #9 S , , #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHC(R26)-T2, R12 is phenyl which may be substituted by methoxycarbonyl, carboxyl or a group of the a –S(O)2OH, R13 is phenyl which may be substituted by methoxycarbonyl or carboxyl, R26 is en or hydroxy, T2 is phenyl, benzyl, 1H-indolyl or 1H-indolylmethyl, R35 is methyl or hydroxy, (followed by page 13A) and also their salts, solvates and solvates of the salts.
In a preferred ment AK is a binder which is an antibody or an antigen-binding antibody fragment that binds to FGFR2.
In another preferred embodiment G is a carbonyl or a group of the formula O O HO #1 #1 #2 O N H N #2 H #1 N #2 O , OH O or #1 where #1 marks the linkage site with the sulphur atom of the binder, #2 marks the linkage site with the group L1, L1 is a bond, linear (C1-C10)-alkanediyl, or a group of the formula ##1 ##2 O m or ##1 L1A B1 L1B ##2 where m is a number from 2 to 6, ## 1 marks the e site with the group G, ##2 marks the linkage site with the group B, (followed by page 13B) L1A is linear (C2-C10)-alkanediyl, B1 is a group of the formula O O O ##5 ##6 L5 ##6 N N ##5 N N L6 R29 R30 , R31 R32 or S ##6 ##5 S in which ## 5 marks the linkage site with the group L1A, ## 6 marks the linkage site with the group L1B, L5 is a bond or (C2-C4)-alkanediyl, L6 is a bond or a group of the formula R33 HN HN O O ##8 or ##8 ##7 ##7 R34 O in which ## 7 marks the linkage site with the carbonyl group, ## 8 marks the linkage site with L1B, R33 is hydrogen, )-alkylcarbonyl, tert-butyloxycarbonyl, or benzyloxycarbonyl, R34 is hydrogen or methyl, wed by page 13C) R29 is hydrogen or (C1-C4)-alkyl, R30 is hydrogen or (C1-C4)-alkyl, R29 and R30 together with the atoms to which they are bonded form a 5- or 6-membered heterocycle, R31 is hydrogen or (C1-C4)-alkyl, R32 is hydrogen or (C1-C4)-alkyl, R31 and R32 together with the atoms to which they are bonded form a 5- or 6-membered heterocycle, L1B is linear (C2-C10)-alkanediyl, where (C2-C10)-alkanediyl is optionally substituted by 1 to 4 substituents selected independently of one another from the group ting of methyl, hydroxy and benzyl, where two carbon atoms of the alkanediyl chain in 1,2, 1,3 or 1,4-relation to one r, with inclusion of any carbon atoms situated between them, are optionally bridged to form a (C3-C6)-cycloalkyl ring or a phenyl ring, B is a bond or a group of the formula (followed by page 13D) O O O O * ** L3 ** O ** N N * N N L4 * Q1 R14 R15 R16 R17 O , , , R20 O * P ** * N ** Q2 * ** O , R18 R19 O , R21 R22 , O O R36 O ** ** or * N * N O ** H * R23 R24 O R27 , R37 O where * marks the linkage site with L1, ** marks the linkage site with L2, P is O or NH, L3 is a bond or )-alkanediyl, L4 is a bond or a group of the formula R28 HN HN O O **** or *** **** R25 O in which (followed by page 13E) *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is hydrogen or methyl, R28 is hydrogen, (C1-C4)-alkylcarbonyl, tert-butyloxycarbonyl, or benzyloxycarbonyl, Q1 is a 4- to 7-membered heterocycle, Q2 is a 3- to 7-membered carbocycle or a 4- to 7-membered heterocycle, R14 is hydrogen or (C1-C4)-alkyl, R15 is hydrogen or (C1-C4)-alkyl, R14 and R15 together with the atoms to which they are bonded form a 5- or 6- membered cycle, R16 is hydrogen or (C1-C4)-alkyl, R17 is hydrogen or (C1-C4)-alkyl, R16 and R17 together with the atoms to which they are bonded form a 5- or 6- ed heterocycle, R18 is hydrogen or (C1-C4)-alkyl, R19 is hydrogen or the side group of a natural o acid or of its homologues or isomers, R20 is hydrogen or (C1-C4)-alkyl, (followed by page 13F) R19 and R20 together with the atoms to which they are bonded form a pyrrolidinyl ring, R21 is hydrogen or (C1-C4)-alkyl, R22 is hydrogen or (C1-C4)-alkyl, R21 and R22 together with the atoms to which they are bonded form a 3- to 7- membered carbocycle, R23 is (C1-C4)-alkyl, R24 is hydrogen or (C1-C4)-alkyl, R27 is hydrogen or (C1-C4)-alkyl, R36 is en, (C1-C4)-alkylcarbonyl, tert-butyloxycarbonyl, or benzyloxycarbonyl R37 is hydrogen or methyl, R36 and R37 together with the atoms to which they are bonded form a pyrrolidine ring, and also their salts, solvates and solvates of the salts. wed by page 14) reproduced in the g examples, by using corresponding isotopic modifications of the tive reagents and/or starting compounds.
Preferred salts in the context of the present invention are physiologically acceptable salts of the nds of the invention. Also encompassed are salts which gh themselves not suitable for pharmaceutical applications may nevertheless be used, for example, for isolating or ing the compounds of the invention.
Physiologically acceptable salts of the compounds of the invention encompass acid addition salts of mineral acids, carboxylic acids and sulphonic acids, examples being salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, ethanesulphonic acid, benzenesulphonic acid, toluenesulphonic acid, naphthalenedisulphonic acid, acetic acid, oroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
Physiologically acceptable salts of the compounds of the invention also encompass salts of customary bases, such as, by way of example and preferably, alkali metal salts (e.g. sodium and potassium salts), alkaline earth metal salts (e.g. calcium and magnesium salts) and ammonium salts, d from ammonia or organic amines having 1 to 16 C atoms, such as, by way of example and preferably, ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, ylaminoethanol, procaine, dibenzylamine, N-methylpiperidine, N-methylmorpholine, arginine, lysine and 1,2- ethylenediamine.
Solvates in the context of the invention are those forms of the compounds of the invention that form a complex in the solid or liquid state through coordination with solvent molecules. es are one ic form of solvates, in which the coordination takes place with water. Preferred solvates in the t of the t invention are hydrates.
Furthermore, the present invention also asses prodrugs of the compounds of the invention.
The term “prodrugs” here identifies compounds which may themselves be biologically active or inactive but are converted during their residence in the body into compounds of the invention (by metabolism or hydrolysis, for example).
In the context of the present ion the definitions of the substituents, unless otherwise specified, are as follows: (C 1-C4)-Alkyl in the context of the invention is a linear or branched alkyl radical having 1 to 4 carbon atoms. By way of example and with preference, the following may be mentioned: methyl, ethyl, n-propyl, isopropyl, l, isobutyl, 1-methylpropyl and tert -butyl.
Alkanediyl in the context of the invention is a linear, α,ω-divalent alkyl radical having the ular number of carbon atoms indicated. By way of example and of preference, the following may be mentioned: methylene, -1,2-diyl (1,2-ethylene), propane-1,3-diyl (1,3-propylene), butane-1,4-diyl (1,4-butylene), pentane-1,5-diyl entylene), hexane-1,6-diyl (1,6-hexylene), heptane-1,7-diyl (1,7-hexylene), octane-1,8-diyl (1,8-octylene), nonane-1,9-diyl (1,9-nonylene), decane-1,10-diyl decylene).
(C 3-C7)-Cycloalkyl and 3- to 7-membered carbocycle respectively in the context of the invention is a clic, saturated cycloalkyl group having 3 to 7 carbon atoms. By way of example and of preference, the following may be mentioned: cyclopropyl, cyclobutyl, entyl, cyclohexyl and cycloheptyl.
The side group of an α-amino acid in the definition of R19 encompasses not only the side groups of the naturally occurring α-amino acids but also the side groups of homologues and isomers of these α-amino acids. The α-amino acid here may be in the L or D configuration or else may be present as a mixture of the L and D forms. Examples that may be given of side groups are as follows: methyl (alanine), propanyl (valine), yl (norvaline), 2-methylpropanyl (leucine), 1-methylpropanyl (isoleucine), 1-yl (norleucine), tert -butyl (2-tert -butylglycine), phenyl (2-phenylglycine), benzyl (phenylalanine), p-hydroxybenzyl (tyrosine), indolylmethyl (tryptophan ), imidazolylmethyl (histidine), hydroxymethyl e), 2-hydroxyethyl (homoserine), 1-hydroxyethyl (threonine), mercaptomethyl (cysteine), methylthiomethyl (S-methylcysteine), 2-mercaptoethyl (homocysteine), 2-methylthioethyl (methionine), carbamoylmethyl (asparagine), 2-carbamoylethyl (glutamine), carboxymethyl (aspartic acid), 2-carboxyethyl (glutamic acid), 4-aminobutanyl (lysine), ohydroxybutanyl (hydroxylysine), 3-aminopropanyl hine), oethyl (2,4-diaminobutyric acid), aminomethyl (2,3-diaminopropionic acid), 3-guanidinopropanyl (arginine), 3-ureidopropanyl (citrulline). Preferred α-amino acid side groups in the definition of R19 are methyl (alanine), propanyl (valine), ylpropanyl ne), benzyl (phenylalanine), olylmethyl (histidine), hydroxymethyl (serine), 1-hydroxyethyl (threonine), 4-aminobutanyl e), 3-aminopropanyl (ornithine), 2-aminoethyl (2,4-diaminobutyric acid), aminomethyl (2,3-diaminopropionic acid), 3-guanidinopropanyl (arginine). The L configuration is preferred in each case.
A 4- to 7-membered heterocycle in the context of the invention is a monocyclic, saturated heterocycle having a total of 4 to 7 ring atoms, which contains one or two ring heteroatoms from the series N, O, S, SO and/or SO2 and is linked via a ring carbon atom or optionally a ring en atom. Preference is given to a 5- to 7-membered heterocycle having one or two ring heteroatoms from the series N, O and/or S, more preferably a 5- or 6-membered heterocycle having one or two ring heteroatoms from the series N and/or O. By way of example, the following may be mentioned: azetidinyl, oxetanyl, pyrrolidinyl, pyrazolidinyl, tetrahydrofuranyl, thiolanyl, piperidinyl, piperazinyl, tetrahydropyranyl, ydrothiopyranyl, linyl, thiomorpholinyl, hexahydroazepinyl and hexahydro-1,4-diazepinyl. ence is given to pyrrolidinyl, tetrahydrofuranyl , piperidinyl, piperazinyl, tetrahydropyranyl and morpholinyl.
In the formula of the group which may be represented by A, B, D, G, L1, L2, L4, R1, R2, R3, R4 and R5, respectively, the end point of the line at which the symbol #6, *, **, #3, #1, #2, ##1, ##2, ##3, ##4, ***, ****, #4, #5, #6, #7, #8 or #9 is located is not a carbon atom or a CH2 group, but instead is part of the bond to the atom designated in each case, to which the A, B, D, G, L1, L2, L4, R1, R2, R3, R4 or R5 is bonded.
In the context of the present invention, all radicals which occur multiply have their definition ndently of one another. If radicals in the compounds of the invention are substituted, the radicals, unless ise specified, may be substituted one or more times. Substitution by one or by two cal or different substituent(s) is preferred. ularly preferred is substitution by one substituent.
In the context of the present invention the terms used, unless otherwise specified, have the ing definitions: The term r” is understood in the st sense as a chemical unit which comprises a covalent bond or a series of atoms that links a binder covalently to a drug. The term “linker” is understood preferably as a series of atoms in the sense of the t invention that links a binder covalently to a drug. Furthermore, linkers may be represented, for example, by divalent chemical units, such as alkyldiyls, aryldiyls, heteroaryldiyls, heterocyclyldiyls, dicarbonyl acid esters, onyl acid amides.
The term “binder” is understood in the broadest sense as a molecule which binds to a target molecule which is present on a particular target cell population to be addressed with the binder- drug conjugate. The term “binder” should be understood in its broadest interpretation and encompasses, for example, s, proteins which are able to bind particular sugar chains, or phospholipid-binding proteins. Such binders comprise, for example, high molecular mass proteins (binding proteins), polypeptides or peptides (binding es), non-peptidic (e.g. aptamers (US5,270,163) review article by Keefe AD., et al., Nat. Rev. Drug Discov. 2010; 9:537-550), or vitamins) and all other cell-binding molecules or substances. Binding proteins are, for example, antibodies and dy fragments or antibody mimetics such as, for example, affibodies, adnectins, anticalins, DARPins, avimers, nanobodies w articles by Gebauer M. et al., Curr.
Opinion in Chem. Biol. 2009; 13:245-255; Nuttall S.D. et al., Curr. Opinion in Pharmacology 2008; 8:608-617). Binding peptides are, for example, ligands of a ligand-receptor pair, such as VEGF in the ligand-receptor pair VEGF/KDR, such as transferrin of the ligand-receptor pair transferrin/transferrin receptor, or cytokines/cytokine receptor, such as TNFalpha in the ligand or pair TNFalpha/TNFalpha receptor.
The term “epitope” as used herein encompasses any determinants of a protein that are able to bind specifically to an immunoglobulin or T-cell receptor. Such determinants commonly consist of chemically active surface arrangements of molecules, such as amino acids, carbohydrates or a ation thereof, for example, which commonly have a specific dimensional structure and also defined charge properties. Two antibodies bind to the same epitope if it is shown in a competitive g assay format that the first antibody es with the second antibody.
Binding assays of this kind are known to the d person.
A “target molecule” is understood in the st sense to be a molecule which is present in the target cell population, and may be a protein (e.g. a receptor of a growth factor) or a non-peptidic molecule (e.g. a sugar or phospholipid). Preferably it is a receptor or an antigen.
The term “extracellular” target molecule describes a target molecule which is attached to the cell and which is located on the outside of a cell or the part of a target molecule which is located on the outside of a cell, i.e. a binder may bind to an intact cell at its extracellular target molecule. An extracellular target molecule may be anchored in the cell membrane or may be part of the cell membrane. The skilled person knows of methods for identifying extracellular target molecules.
For proteins this may be done via determination of the transmembrane domain(s) and the orientation of the protein in the membrane. This data is generally recorded in protein databases (e.g. SwissProt).
The term “cancer target molecule” describes a target molecule which is multiply present on one or more cancer cell types in comparison to non-cancer cells of the same tissue type. The cancer target molecule is ably present selectively on one or more cancer cell types in comparison to non- cancer cells of the same tissue type, with “selectively” describing an at least twofold accumulation on cancer cells in ison to non-cancer cells of the same tissue type (a “selective cancer target molecule”). The use of cancer target molecules allows selective therapy of cancer cells with the conjugates of the invention.
The binder may be linked via a bond to the linker. The g of the binder may take place by means of a heteroatom of the binder. Inventive heteroatoms of the binder that may be used for linking are sulphur (in one embodiment via a sulphhydryl group of the binder), oxygen (in accordance with the invention by means of a carboxyl or hydroxy group of the binder) and nitrogen (in one embodiment via a primary or ary amine group or amide group of the binder). These heteroatoms may be present in the natural binder or may be introduced by means of methods of chemistry or molecular biology. In accordance with the invention, the linking of the binder to the toxophore has little influence over the binding activity of the binder to the target molecule. In a red embodiment the linking has no influence on the binding activity of the binder to the target molecule.
The term “antibody” is understood in accordance with the present invention in its broadest sense and encompasses immunoglobulin molecules, examples being intact or modified monoclonal antibodies, polyclonal antibodies or multispecific antibodies (e.g. bispecific antibodies). An immunoglobulin molecule preferably comprises a molecule having four polypeptide chains, two heavy chains (H chains) and two light chains (L chains), which are linked typically by disulphide bridges. Each heavy chain comprises a le domain of the heavy chain (abbreviated to VH) and a constant domain of the heavy chain. The constant domain of the heavy chain may encompass, for example, three domains CH1, CH2 and CH3. Each light chain comprises a variable domain (abbreviated to VL) and a nt domain. The constant domain of the light chain comprises one domain (abbreviated to CL). The VH and VL domains may be further subdivided into regions having hypervariability, also called complementarity-determining s viated to CDR), and regions having a low sequence ility ework region”, abbreviated to FR).
Each VH and VL region is typically composed of three CDRs and up to four FRs. For example, in the following order from the amino terminus to the carboxy us: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. An antibody may be obtained from any species suitable for the antibody, such as, for example, rabbit, llama, camel, mouse or rat. In one embodiment the antibody is of human or murine origin. An antibody may for example be human, humanized or chimeric.
The term lonal” antibody identifies antibodies which have been obtained from a population of ntially neous antibodies, i.e. individual antibodies of the population are identical except for naturally ing mutations which may occur in small numbers. Monoclonal dies recognize a single antigenic binding site with a high specificity. The term “monoclonal antibody” does not refer to a particular production method.
The term “intact” antibody refers to antibodies which comprise not only an antigen-binding domain but also the constant domain of the light and heavy chain. The constant domain may be a naturally occurring , or a variant thereof in which a plurality of amino acid positions have been altered.
The term “modified ” antibody refers to intact antibodies which have been fused with another polypeptide or protein, not originating from an antibody, via the amino terminus or carboxyl terminus thereof, by means of a covalent bond (e.g. a peptide linkage). rmore, dies may be modified by introducing reactive cysteines at defined locations, in order to facilitate coupling to a toxophore (see Junutula et al. Nat Biotechnol. 2008 Aug;26(8):925-32). 2012/075277 The term “human” dy identifies antibodies which can be obtained from a human being or are synthetic human antibodies. A etic” human antibody is an antibody which in parts or as a whole is obtainable from tic sequences in silico which are based on the analysis of human antibody sequences. A human antibody may be encoded, for example, by a nucleic acid which has been isolated from a library of antibody sequences which are of human . One example of such antibodies can be found in Söderlind et al., Nature Biotech. 2000, 18:853-856.
The term “humanized” or “chimeric” antibody describes antibodies which consist of a non-human and of a human sequence component. In these antibodies, part of the sequences of the human immunoglobulin (recipient) is replaced by sequence components of a non-human immunoglobulin (donor). In many cases the donor is a murine immunoglobulin. With humanized antibodies, amino acids of the CDR in the ent are replaced by amino acids of the donor. In some cases, amino acids of the framework as well are replaced by corresponding amino acids of the donor. In some cases the humanized antibody contains amino acids which were present neither in the ent nor in the donor and which were inserted during the optimization of the antibody. In the case of chimeric antibodies, the le domains of the donor immunoglobulin are fused with the constant regions of a human antibody.
The term complementarity-determining region (CDR) as used here refers to those amino acids in a variable antibody domain that are necessary for binding to the n. Every variable region typically has three CDR regions, identified as CDR1, CDR2 and CDR3. Each CDR region may comprise amino acids according to the definition of Kabat and/or amino acids of a hypervariable loop, defined according to Chotia. The tion according to Kabat encompasses, for example, the region of approximately amino acid position 24 – 34 (CDR1), 50 – 56 (CDR2) and 89 – 97 (CDR3) of the variable light chain and 31 – 35 (CDR1), 50 – 65 (CDR2) and 95 – 102 (CDR3) of the variable heavy chain (Kabat et al., Sequences of Proteins of Immulological Interest, 5th Ed.
Public Health e, National Institutes of , Bethesda, MD. (1991)). The definition according to Chotia encompasses, for example, the region of approximately amino acid position 26 – 32 , 50 – 52 (CDR2) and 91 – 96 (CDR3) of the variable light chain and 26 – 32 (CDR1), 53 – 55 (CDR2) and 96 – 101 (CDR3) of the variable heavy chain Chothia and Lesk; J Mol Biol 196: 901-917 (1987)). In some cases a CDR may comprise amino acids from one CDR region as defined by Kabat and Chotia.
Depending on the amino acid sequence of the constant domain of the heavy chain, antibodies may be d into different classes. There are five main classes of intact antibodies: IgA, IgD, IgE, IgG and IgM, and a number of them may be broken down into further subclasses (isotypes), e.g.
IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2. The constant domains of the heavy chain that correspond to the different classes are identified as [alpha/α], [delta/δ], [epsilon/ε], [gamma/γ] and [mu/ µ].
Both the three-dimensional structure and the t ure of antibodies are known.
The term ional fragment” or “antigen-binding antibody fragments” of a dy/immunoglobulin is defined as a fragment of an antibody/immunoglobulin (e.g. the variable domains of an IgG) which further encompasses the antigen binding domains of the antibody/immunoglobulin. The “antigen binding domain” of an antibody typically encompasses one or more hypervariable regions of an antibody, e.g. the CDR, CDR2 and/or CDR3 region. r, the work” or old” region of an antibody may also play a part with regard to the binding of the antibody to the antigen. The framework region forms the scaffold for the CDRs.
The antigen-binding domain preferably encompasses at least amino acids 4 to 103 of the variable light chain and amino acid 5 to 109 of the variable heavy chain, more preferably amino acid 3 to 107 of the le light chain and 4 to 111 of the variable heavy chain, particular preference being given to the complete variable light and heavy chains, i.e. amino acid 1 – 109 of the VL and 1 to 113 of the VH (numbering according to 8320).
“Functional fragments” or “antigen-binding antibody fragments” of the invention encompass, nonconclusively , Fab, Fab’, F(ab’)2 and Fv fragments, diabodies, Single Domain Antibodies (DAbs), linear antibodies, individual chains of antibodies (single-chain Fv, abbreviated to ScFv); and multispecific antibodies, such as bi and tri-specific antibodies, for example, formed from antibody nts C. A. K Borrebaeck, editor (1995) Antibody Engineering (Breakthroughs in Molecular Biology), Oxford University Press; R. Kontermann & S. Duebel, editors (2001) Antibody Engineering (Springer Laboratory Manual), Springer Verlag). Antibodies other than “multispecific” or “multifunctional” dies are those having identical binding sites.
Multispecific antibodies may be specific for different epitopes of an antigen or may be specific for epitopes of more than one antigen (see, for example WO 93/17715; WO 92/08802; WO 60; WO 92/05793; Tutt, et al., 1991, J. Immunol. 147:60 69; U. S. Pat. Nos. 4,474,893; 4,7 14,68 1; 4,925,648; 920; 5,601,8 19; or Kostelny et al., 1992, J. Immunol. 148: 1547 1553). An F(ab’) 2 or Fab molecule may be constructed such that the number of intermolecular disulphide interactions occurring between the Ch1 and the CL domains can be reduced or else completely prevented.
“Functional fragments” or “antigen-binding antibody fragments” may be fused with another polypeptide or protein, not originating from an dy, via the amino terminus or carboxyl terminus thereof, by means of a covalent bond (e.g. a e linkage). Furthermore, antibodies and antigen-binding fragments may be modified by introducing reactive cysteines at defined locations, in order to facilitate coupling to a toxophore (see la et al. Nat Biotechnol. 2008 Aug;26(8):925-32). 2012/075277 Polyclonal dies can be prepared by methods known to a person of ordinary skill in the art.
Monoclonal antibodies may be prepared by methods known to a person of ordinary skill in the art (Köhler and Milstein, Nature, 256, 495-497, 1975). Human and humanized onal antibodies may be prepared by methods known to a person of ordinary skill in the art (Olsson et al., Meth Enzymol. 92, 3-16 or Cabilly et al US 567 or Boss et al US 4,816,397).
A person of ordinary skill in the art is aware of diverse methods for preparing human antibodies and fragments thereof, such as, for example, by means of transgenic mice (N Lonberg and D Huszar, Int Rev Immunol. 1995;13(1):65-93) or Phage Display Technologies (Clackson et al., Nature. 1991 Aug 15;352(6336):624-8). Antibodies of the invention may be obtained from recombinant antibody libraries consisting for example of the amino acid sequences of a multiplicity of dies compiled from a large number of healthy volunteers. Antibodies may also be produced by means of known recombinant DNA technologies. The nucleic acid sequence of an antibody can be obtained by routine sequencing or is available from publically accessible databases.
An “isolated” antibody or binder has been purified to remove other constituents of the cell.
Contaminating constituents of a cell which may interfere with a stic or therapeutic use are, for example, enzymes, hormones, or other peptidic or ptidic constituents of the cell. A preferred antibody or binder is one which has been purified to an extent of more than 95%, relative to the antibody or binder mined for example by Lowry method, UV-Vis spectroscopy or by SDS ary gel electrophoresis). Furthermore an antibody which has been ed to such an extent that it is possible to determine at least 15 amino acids of the amino terminus or of an al amino acid sequence, or which has been purified to homogeneity, the homogeneity being determined by SDS-PAGE under reducing or non-reducing conditions (detection may be determined by means of Coomassie Blau staining or preferably by silver tion). However, an antibody is normally prepared by one or more purification steps.
The term “specific binding” or “binds specifically” refers to an antibody or binder which binds to a predetermined antigen/target le. Specific binding of an antibody or binder typically describes an antibody or binder having an affinity of at least 10-7 M, with the antibody or binder having an at least two times higher affinity for the ermined antigen/target molecule than for a non-specific n/target molecule (e.g. bovine serum albumin, or casein) which is not the ermined antigen/target molecule or a closely related antigen/target molecule.
Antibodies which are specific t a cancer cell antigen can be prepared by a person of ordinary skill in the art by means of methods with which he or she is familiar (such as recombinant expression, for example) or may be acquired commercially (as for example from Merck KGaA, Germany). Examples of known commercially available antibodies in cancer therapy are Erbitux® (cetuximab, Merck KGaA), Avastin® (bevacizumab, Roche) and Herceptin® (trastuzumab, Genentech). Trastuzumab is a recombinant humanized monoclonal antibody of the IgG1kappa type which in a cell-based assay (Kd = 5 nM) binds the ellular domains of the human epidermal growth receptor with high affinity. The antibody is produced inantly in CHO cells.
A preferred subject of the invention are binder-drug ates of the general formula (Ia) in which n is a number from 1 to 50, AK is AK1 or AK2 where AK1 is a binder which binds FGFR2 and is bonded via a sulphur atom of the binder to the group G, AK2 is a binder which binds FGFR2 and is bonded via a nitrogen atom of the binder to the group G, G when AK = AK1, is a group of the formula O O HO #1 #1 #2 O N H N #2 H #1 N #2 O , OH O or #1 O where #1 marks the linkage site with the sulphur atom of the binder, #2 marks the linkage site with the group L1, when AK = AK2, is carbonyl, L1 is a bond, linear (C1-C10)-alkanediyl, a group of the formula ##1 ##2 O m or ##1 L1A B1 L1B ##2 where m is a number from 2 to 6, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, L1A is linear (C2-C10)-alkanediyl, B1 is a group of the formula O O O ##5 ##6 L5 ##6 N N ##5 N N L6 R29 R30 , R31 R32 or S ##6 ##5 S in which ## 5 marks the linkage site with the group L1A, ## 6 marks the e site with the group L1B, L5 is a bond or (C2-C4)-alkanediyl, L6 is a bond or a group of the formula R33 HN HN O O ##8 or ##8 ##7 ##7 R34 O in which ## 7 marks the e site with the carbonyl group, ## 8 marks the linkage site with L1B , R33 is hydrogen, (C1-C4)-alkylcarbonyl, tert-butyloxycarbonyl or benzyloxycarbonyl, R34 is hydrogen or methyl, R29 is hydrogen or (C1-C4)-alkyl, R30 is hydrogen or (C1-C4)-alkyl, R29 and R30 together with the atoms to which they are bonded form a 5- or 6- membered heterocycle, R31 is hydrogen or (C1-C4)-alkyl, R32 is hydrogen or (C1-C4)-alkyl, R31 and R32 together with the atoms to which they are bonded form a 5- or 6- membered heterocycle, L1B is linear 0 )-alkanediyl, where (C1-C10 )-alkanediyl may be substituted by 1 to 4 substituents selected independently of one another from the group consisting of , hydroxy and benzyl, where two carbon atoms of the diyl chain in 1,2, 1,3 or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, may be bridged to form a (C3-C6)-cycloalkyl ring or a phenyl ring, B is a bond or a group of the formula O O O O * ** L3 ** O ** N N * N N L4 * Q1 R14 R15 R16 R17 O , , , R20 O * P ** * N ** Q2 * ** O , R18 R19 O , R21 R22 , O O R36 O ** ** or N * N O ** H * R23 R24 O R27 , R37 O where * marks the linkage site with L1, ** marks the linkage site with L2, P is O or NH, L3 is a bond or (C2-C4)-alkanediyl, L4 is a bond or a group of the formula R28 HN HN O O **** or *** **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is en or methyl, R28 is en, (C1-C4)-alkylcarbonyl, utyloxycarbonyl or benzyloxycarbonyl, Q1 is a 4- to 7-membered heterocycle, Q2 is a 3- to 7-membered carbocycle or a 4- to 7-membered heterocycle, R14 is hydrogen or (C1-C4)-alkyl, R15 is hydrogen or (C1-C4)-alkyl, R14 and R15 together with the atoms to which they are bonded form a 5- or 6-membered heterocycle, R16 is hydrogen or (C1-C4)-alkyl, R17 is hydrogen or (C1-C4)-alkyl, R16 and R17 together with the atoms to which they are bonded form a 5- or 6-membered cycle, R18 is hydrogen or (C1-C4)-alkyl, R19 is hydrogen or the side group of a natural α-amino acid or of its homologues or isomers, R20 is hydrogen or )-alkyl, R19 and R20 together with the atoms to which they are bonded form a pyrrolidinyl ring, R21 is hydrogen or (C1-C4)-alkyl, R22 is hydrogen or (C1-C4)-alkyl, R21 and R22 together with the atoms to which they are bonded form a 3- to 7-membered carbocycle, R23 is (C1-C4)-alkyl, R24 is hydrogen or (C1-C4)-alkyl, R27 is hydrogen or (C1-C4)-alkyl, R36 is hydrogen, (C1-C4)-alkylcarbonyl, tert-butyloxycarbonyl or benzyloxycarbonyl, R37 is en or methyl, R36 and R37 together with the atoms to which they are bonded form a idine ring, L2 is linear (C2-C10)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, where (C2-C10)-alkanediyl may be substituted by 1 to 4 substituents selected independently of one another from the group consisting of methyl, hydroxy and benzyl, where two carbon atoms of the alkanediyl chain in 1,2, 1,3 or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, may be d to form a (C3-C6)-cycloalkyl ring or a phenyl ring, D is a group of the formula O #3 R5 #3 T1 N or #3 R3 R4 O , R26 T2 R1 R2 in which #3 marks the e site with the nitrogen atom, R1 is hydrogen or methyl, R2 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, 1-hydroxyethyl, 4- ybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, 1-phenylethyl , diphenylmethyl, 1H-imidazolylmethyl or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a ) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula O O O or N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the yl group, R6 is hydrogen, hydroxy or benzyloxy, R3 is hydrogen or methyl, R4 is isopropyl, isobutyl, sec-butyl, utyl, phenyl, benzyl, 1-hydroxyethyl, 4- hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, ylethyl , diphenylmethyl, 1H-imidazolylmethyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9, -C(=O)-NH-NH-R10 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen or methyl, R9 is hydrogen, , ethyl, n-propyl or benzyl, or R8 and R9 together with the en atom to which they are bonded form a 4- to ered heterocycle, R10 is benzoyl, R11 is benzyl, which may be substituted in the phenyl group by methoxycarbonyl or carboxyl, R5 is hydrogen, methyl or a group of the formula #9 O #9 S , , #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHC(R26)-T2, R12 is phenyl which may be substituted by methoxycarbonyl, yl or a group of the formula –S(O)2OH, R13 is phenyl which may be substituted by methoxycarbonyl or carboxyl, R26 is hydrogen or hydroxy, T2 is phenyl, benzyl, 1H-indolyl or 1H-indolylmethyl, R35 is methyl or hydroxy, and also their salts, solvates and solvates of the salts.
Preferred subject of the invention are binder-drug conjugates of the general formula (Ia) in which n is a number from 1 to 20, AK is AK1 or AK2 where AK1 is a binder which binds FGFR2 and is bonded via the sulphur atom of a ne residue of the binder to the group G, AK2 is a binder which binds FGFR2 and is bonded via the NH side group of a lysine residue of the binder to the group G, G when AK = AK1, is a group of the formula N #2 in which #1 marks the linkage site with the cysteine e of the binder, #2 marks the linkage site with the group L1, or when AK = AK2, is carbonyl, L1 is a bond, linear (C2-C6)-alkanediyl, a group of the formula ##1 ##2 O m or ##1 L1A B1 L1B ##2 where m is a number from 2 to 6, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, L1A is linear (C2-C6)-alkanediyl, B1 is a group of the formula O O O ##5 ##6 L5 ##6 N N ##5 N N L6 R29 R30 , R31 R32 or S ##6 ##5 S in which ## 5 marks the linkage site with the group L1A, ## 6 marks the linkage site with the group L1B, L5 is a bond, L6 is a bond or a group of the formula R33 HN HN O O ##8 or ##8 ##7 ##7 R34 O in which ## 7 marks the linkage site with the carbonyl group, ## 8 marks the linkage site with L1B, R33 is hydrogen, carbonyl or tert-butyloxycarbonyl, R34 is hydrogen or methyl, R29 is hydrogen, R30 is hydrogen, R31 is hydrogen or methyl, R32 is hydrogen or methyl, L1B is linear (C2-C6)-alkanediyl, and where (C2-C6)-alkanediyl may be substituted by 1 or 2 methyl substituents, B is a bond or a group of the a O O O O * ** L3 ** O ** N N * N N L4 * Q1 R14 R15 R16 R17 O , , , R20 O O * N ** ** * ** O N * N H H R18 R19 O , R21 R22 , R23 R24 O O R36 ** or * N O ** R37 O where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a bond or a group of the formula R28 HN HN O O **** or *** **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is hydrogen or methyl, R28 is en, methylcarbonyl or tert-butyloxycarbonyl, Q1 is a 4- to 7-membered heterocycle, R14 is hydrogen, R15 is hydrogen, R16 is hydrogen or , R17 is hydrogen or methyl, R16 and R17 together with the atoms to which they are bonded form a piperazinyl ring, R18 is hydrogen, R19 is hydrogen, methyl, propanyl , ylpropanyl or 1-methylpropanyl, R20 is hydrogen or methyl, R19 and R20 together with the atoms to which they are bonded form a pyrrolidinyl ring, R21 is hydrogen or methyl, R22 is hydrogen or methyl, R21 and R22 together with the atoms to which they are bonded form a cyclopropyl ring, R23 is methyl, R24 is hydrogen or methyl, R27 is hydrogen, R36 is hydrogen, methylcarbonyl or tert-butyloxycarbonyl, R37 is hydrogen or methyl, R36 and R37 together with the atoms to which they are bonded form a pyrrolidine ring, L2 is linear (C2-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, ## 3 marks the linkage site with the group B, ##4 marks the e site with the en atom, where (C2-C10)-alkanediyl may be substituted by 1 or 2 methyl substituents, D is a group of the formula O #3 R5 #3 T1 N or #3 R3 R4 O , R26 T2 R1 R2 where 2012/075277 #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is oxyethyl, benzyl, 4-hydroxybenzyl, 1-phenylethyl or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally tuted heterocycle of the formula O O or O N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy or benzyloxy, R3 is hydrogen, R4 is 1-hydroxyethyl, benzyl, 4-hydroxybenzyl, 1-phenylethyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent en atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -NR8R9, -C(=O)-NH-NH-R10 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen or methyl, R9 is en, methyl, ethyl, n-propyl or benzyl, R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to 7-membered heterocycle, R10 is benzoyl, R11 is , which may be substituted in the phenyl group by ycarbonyl or carboxyl, R5 is hydrogen, methyl or a group of the formula #9 O #9 S , , #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHC(R26)-T2, R12 is phenyl which may be substituted by methoxycarbonyl, carboxyl or a group of the formula –S(O)2OH, R13 is phenyl which may be substituted by methoxycarbonyl or carboxyl, R26 is hydrogen or hydroxy, T2 is phenyl, benzyl, 1H-indolyl or 1H-indolylmethyl, R35 is methyl or hydroxy, and also their salts, solvates and solvates of the salts.
Preferred subject matter of the invention are -drug conjugates of the general formula (Ia), in which n is a number from 1 to 10, AK is AK1 or AK2 where AK1 is a binder which binds FGFR2 and is bonded via the sulphur atom of a cysteine residue of the binder to the group G, AK2 is a binder which binds FGFR2 and is bonded via the NH side group of a lysine residue of the binder to the group G, G when AK = AK1, is a group of the formula N #2 in which #1 marks the linkage site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, when AK = AK2, is carbonyl, L1 is a bond, linear (C2-C6)-alkanediyl, a group of the a ##1 ##2 O m where m is a number 2 or 3, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C2-C6)-alkanediyl may be substituted by 1 or 2 methyl substituents, B is a bond or a group of the formula O O O L3 ** O ** * N N L4 * Q1 R16 R17 , O , O O * ** or O ** N * N H H R21 R22 R23 R24 O where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a bond or a group of the formula R28 HN HN O O **** or *** **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is methyl, R28 is en, methylcarbonyl or tert-butyloxycarbonyl, Q1 is piperidine-1,4-diyl, R16 is hydrogen or methyl, R17 is hydrogen or methyl, 2012/075277 R16 and R17 together with the atoms to which they are bonded form a piperazinyl ring, R21 is hydrogen or methyl, R22 is hydrogen or , or R21 and R22 together with the atoms to which they are bonded form a cyclopropyl ring, R23 is methyl, R24 is hydrogen, L2 is linear (C2-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, D is a group of the formula O #3 T1 #3 R5 N or #3 R3 R4 O , R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is 1-hydroxyethyl, benzyl, 4-hydroxybenzyl, 1-phenylethyl or 1H-indolylmethyl, 2012/075277 R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present n is a mono- or bicyclic, optionally substituted heterocycle of the formula O O O or O N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy or benzyloxy, R3 is hydrogen, R4 is benzyl, 4-hydroxybenzyl, ylethyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula WO 87716 #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or tylmethyl, R8 is hydrogen or methyl, R9 is hydrogen, methyl, ethyl, n-propyl or benzyl, R11 is , which may be substituted in the phenyl group by methoxycarbonyl or carboxyl, R5 is hydrogen, methyl or a group of the formula #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHCH2phenyl, R12 is phenyl which may be substituted by methoxycarbonyl, carboxyl or a group of the formula –S(O)2OH, R13 is phenyl which may be substituted by methoxycarbonyl or carboxyl, R35 is methyl or hydroxy, and also their salts, es and solvates of the salts.
Preferred subject matter of the present invention are -drug conjugates of the general formula (Ia), in which n is a number from 1 to 10, AK is AK2, where AK2 is a binder which binds FGFR2 and is bonded via the NH side group of a lysine residue of the binder to the group G, G is carbonyl, L1 is a bond, B is a bond, L2 is linear (C3-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, D is a group of the formula O #3 T1 N or #3 R3 R4 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is benzyl, 4-hydroxybenzyl or olylmethyl, R1 and R2 er with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or ic, optionally substituted heterocycle of the formula in which #6 marks the linkage site with the carbonyl group, R3 is hydrogen, R4 is benzyl, 4-hydroxybenzyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the e site with the group T1, T1 is a group of the a -C(=O)-OR7 or -C(=O)-NR8R9 in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen, R9 is hydrogen or benzyl, R35 is methyl, and also their salts, solvates and es of the salts.
Preferred subject matter of the invention are binder-drug conjugates of the general formula (Ia), in which n is a number from 1 to 10, AK is AK1, where AK1 is a binder which binds FGFR2 and is bonded via the sulphur atom of a cysteine residue of the binder to the group G, G is a group of the formula N #2 where #1 marks the linkage site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, L1 is a bond, linear (C3-C5)-alkanediyl or a group of the formula ##1 ##2 O m where m is a number 2 or 3, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C3-C5)-alkanediyl may be substituted by 1 or 2 methyl substituents, B is a bond or a group of the a O O L3 ** * N N L4 R16 R17 where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a bond or a group of the formula O **** R25 O in which *** marks the linkage site with the yl group, **** marks the e site with L2, R25 is methyl, R28 is hydrogen, methylcarbonyl or tert-butyloxycarbonyl, R16 is hydrogen or methyl, R17 is hydrogen or methyl, R16 and R17 together with the atoms to which they are bonded form a piperazinyl ring, L2 is linear (C3-C5)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, D is a group of the formula O #3 T1 N or #3 R3 R4 R1 R2 where #3 marks the linkage site with the en atom, R1 is hydrogen, R2 is benzyl, oxybenzyl or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula in which #6 marks the linkage site with the carbonyl group, R3 is hydrogen, R4 is benzyl, 4-hydroxybenzyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7 or -C(=O)-NR8R9, in which R7 is en, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen, R9 is hydrogen or benzyl, R35 is , and also their salts, solvates and solvates of the salts.
Additionally preferably ed by the present invention is a binder-drug conjugate as bed above, where the binder comprises the amino acid sequence of the variable light and heavy chains of the antibody M048-D01-hIgG1-b, reproduced in SEQ ID NO: 14 (Vl) and SEQ ID NO: 13 (Vh), the amino acid ce of the light and heavy chain of the antibody M048-D01-hIgG1-b reproduced in SEQ ID NO: 9 (light chain) and SEQ ID NO: 10 (heavy chain).
Additionally provided by the present invention are compounds of the formula (XXXa) O H C R35 H C 3 CH 3 3 Cys O H H N N N D N L1 B L2 N N O O CH O CH O O 3 3 CH H CH CH O C 3 3 3 (XXXa), in which Cys is a cysteine residue which is bonded via the sulphur atom of the side chain to a carbon atom of the succinimide, L1 is a bond, linear (C1-C10)-alkanediyl, a group of the formula ##1 ##2 O m or ##1 L1A B1 L1B ##2 where m is a number from 2 to 6, ## 1 marks the linkage site with the group G, ##2 marks the e site with the group B, L1A is linear (C2-C10)-alkanediyl, B1 is a group of the formula O O O ##5 ##6 L5 ##6 N N or ##5 N N L6 R29 R30 R31 R32 in which ## 5 marks the linkage site with the group L1A, 2012/075277 ## 6 marks the linkage site with the group L1B , L5 is a bond or (C2-C4)-alkanediyl, L6 is a bond, R29 is hydrogen or (C1-C4)-alkyl, R30 is hydrogen or (C1-C4)-alkyl, R29 and R30 together with the atoms to which they are bonded form a 5- or 6- membered cycle, R31 is hydrogen or (C1-C4)-alkyl, R32 is hydrogen or (C1-C4)-alkyl, R31 and R32 together with the atoms to which they are bonded form a 5- or 6- membered heterocycle, L1B is linear (C2-C10 )-alkanediyl, and where (C1-C10 )-alkanediyl may be substituted by 1 to 4 substituents selected independently of one another from the group consisting of , hydroxy and benzyl, where two carbon atoms of the alkanediyl chain in 1,2, 1,3 or 1,4-relation to one r, with inclusion of any carbon atoms situated between them, may be bridged to form a (C3- C6)-cycloalkyl ring or a phenyl ring, B is a bond or a group of the formula 2012/075277 O O O * ** L3 ** N N * N N L4 R14 R15 R16 R17 , , O O O ** ** * Q1 or * N O R27 where * marks the linkage site with L1, ** marks the linkage site with L2, P is O or NH, L3 is a bond or (C2-C4)-alkanediyl, L4 is a bond, Q1 is a 4- to 7-membered heterocycle, Q2 is a 3- to 7-membered carbocycle or a 4- to 7-membered heterocycle, R14 is en or (C1-C4)-alkyl, R15 is hydrogen or (C1-C4)-alkyl, R14 and R15 together with the atoms to which they are bonded form a 5- or 6-membered heterocycle, R16 is hydrogen or (C1-C4)-alkyl, R17 is hydrogen or (C1-C4)-alkyl, R16 and R17 together with the atoms to which they are bonded form a 5- or 6-membered heterocycle, R18 is hydrogen or (C1-C4)-alkyl, R19 is hydrogen or the side group of a natural α-amino acid or of its gues or isomers, R20 is hydrogen or (C1-C4)-alkyl, or R19 and R20 together with the atoms to which they are bonded form a pyrrolidinyl ring, R21 is hydrogen or (C1-C4)-alkyl, R22 is hydrogen or (C1-C4)-alkyl, R21 and R22 together with the atoms to which they are bonded form a 3- to 7-membered carbocycle, R23 is (C1-C4)-alkyl, R24 is hydrogen or (C1-C4)-alkyl, R27 is hydrogen or (C1-C4)-alkyl, L2 is linear (C2-C10)-alkanediyl or is a group of the a ##3 ##4 O p where p is a number from 2 to 6, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, where (C2-C10)-alkanediyl may be substituted by 1 to 4 tuents selected ndently of one another from the group consisting of methyl, hydroxy and benzyl, where two carbon atoms of the alkanediyl chain in 1,2, 1,3 or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, may be bridged to form a (C3- C6)-cycloalkyl ring or a phenyl ring, D is a group of the formula O T1 #3 R5 N or #3 R3 R4 O , R26 T2 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen or methyl, R2 is isopropyl, isobutyl, sec-butyl, utyl, phenyl, benzyl, 1-hydroxyethyl, 4- hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, 1-phenylethyl , diphenylmethyl, 1H-imidazolylmethyl or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a ) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the e site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted cycle of the formula O O O or N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy or benzyloxy, R3 is hydrogen or methyl, R4 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, oxyethyl, 4- hydroxybenzyl, oxynitrobenzyl, 4-hydroxyaminobenzyl, 1-phenylethyl , diphenylmethyl, 1H-imidazolylmethyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the e site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9, -C(=O)-NH-NH-R10 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen or , R9 is hydrogen, methyl, ethyl, n-propyl or benzyl, R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to 7-membered heterocycle, R10 is benzoyl, R11 is benzyl, which may be substituted in the phenyl group by methoxycarbonyl or carboxyl, R5 is en, methyl or a group of the formula #9 O #9 S , , #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHC(R26)-T2, R12 is phenyl which may be substituted by ycarbonyl, carboxyl or a group of the a –S(O)2OH, R13 is phenyl which may be substituted by methoxycarbonyl or carboxyl, R26 is hydrogen or hydroxy, T2 is phenyl, benzyl, 1H-indolyl or 1H-indolylmethyl, R35 is methyl or hydroxy, 2012/075277 and also their salts, solvates and solvates of the salts.
Preferred in the context of the present ion are additionally also compounds of the formula (XXXa) in which Cys is a cysteine residue which is bonded via the sulphur atom of the side chain via a carbon atom of the succinimide, L1 is a bond, linear (C2-C6)-alkanediyl, a group of the formula ##1 ##2 O m or ##1 L1A B1 L1B ##2 where m is a number 2 or 3, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, L1A is linear (C2-C6)-alkanediyl, B1 is a group of the formula O O O ##5 ##6 L5 ##6 N N or ##5 N N L6 R29 R30 R31 R32 in which ## 5 marks the linkage site with the group L1A, ## 6 marks the linkage site with the group L1B, L5 is a bond, L6 is a bond, R29 is hydrogen, R30 is hydrogen, R31 is hydrogen or methyl, R32 is hydrogen or methyl, L1B is linear (C2-C6)-alkanediyl, where (C2-C6)-alkanediyl may be substituted by 1 or 2 methyl substituents, B is a bond or a group of the formula O O O * ** L3 ** N N * N N L4 R14 R15 R16 R17 O ** or * N R23 R24 O where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or -1,2-diyl, L4 is a bond, R14 is en, R15 is hydrogen, R16 is hydrogen or methyl, R17 is hydrogen or methyl, R16 and R17 together with the atoms to which they are bonded form piperazinyl ring, WO 87716 R23 is methyl, R24 is hydrogen or methyl, L2 is linear (C2-C6)-alkanediyl or is a group of the a ##3 ##4 O p where p is a number 2 or 3, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, D is a group of the formula O #3 T1 #3 R5 N or #3 R3 R4 O , R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is 1-hydroxyethyl, benzyl, 4-hydroxybenzyl, 1-phenylethyl or 1H-indolylmethyl, or R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, ally substituted heterocycle of the formula O O O or N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy or benzyloxy, R3 is hydrogen, R4 is 1-hydroxyethyl, benzyl, 4-hydroxybenzyl, 1-phenylethyl or olylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula 2012/075277 #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9, -C(=O)-NH-NH-R10 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen or , R9 is hydrogen, methyl, ethyl, n-propyl or , R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to 7- membered heterocycle, R10 is benzoyl, R11 is benzyl which may be substituted in the phenyl group by methoxycarbonyl or carboxyl, R5 is hydrogen, methyl or a group of the formula #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHCH2phenyl, R12 is phenyl which may be tuted by methoxycarbonyl, carboxyl or a group of the formula –S(O)2OH, R13 is phenyl which may be substituted by methoxycarbonyl or carboxyl, R35 is methyl or hydroxy, and also their salts, solvates and solvates of the salts.
Particularly preferred in the context of the present invention are additionally also compounds of the formula (XXXa) in which Cys is a cysteine residue which is bonded via the sulphur atom of the side chain via a carbon atom of the succinimide, L1 is a bond or linear (C2-C6)-alkanediyl, B is a bond or a group of the formula O O L3 ** * N N L4 R16 R17 where * marks the linkage site with L1, ** marks the e site with L2, L3 is a bond, L4 is a bond, R16 is hydrogen or methyl, R17 is hydrogen or , L2 is linear (C2-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, D is a group of the formula O #3 T1 N or #3 R3 R4 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is en, R2 is benzyl or 1H-indolylmethyl, or R1 and R2 together with the carbon atom to which they are bonded form a ) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula 2012/075277 in which #6 marks the linkage site with the carbonyl group, R3 is hydrogen, R4 is benzyl, 4-hydroxybenzyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7 or -C(=O)-NR8R9, in which R7 is hydrogen, R8 is hydrogen, R9 is en, R35 is methyl, and also their salts, solvates and solvates of the salts.
The present invention additionally provides compounds of the a (XXXI) HO NH O 3 O H C R35 H C 3 CH 3 O 3 H N H N H N N D L1 B L2 N N O O CH O CH O O CH 3 3 3 H C CH CH 3 3 (XXXI), in which L1 is a bond, linear (C1-C10)-alkanediyl, a group of the formula ##1 ##2 O m or ##1 L1A B1 L1B ##2 where m is a number from 2 to 6, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, L1A is linear (C2-C10)-alkanediyl, B1 is a group of the formula O O O ##5 ##6 L5 ##6 N N or ##5 N N L6 R29 R30 R31 R32 in which ## 5 marks the linkage site with the group L1A, ## 6 marks the linkage site with the group L1B, L5 is a bond or (C2-C4)-alkanediyl, L6 is a bond, R29 is hydrogen or (C1-C4)-alkyl, R30 is hydrogen or (C1-C4)-alkyl, R29 and R30 together with the atoms to which they are bonded form a 5- or 6- membered heterocycle, R31 is hydrogen or (C1-C4)-alkyl, R32 is hydrogen or (C1-C4)-alkyl, R31 and R32 together with the atoms to which they are bonded form a 5- or 6- ed heterocycle, L1B is linear (C2-C10 )-alkanediyl, where (C1-C10 )-alkanediyl may be substituted by 1 to 4 substituents selected independently of one another from the group consisting of methyl, hydroxy and , where two carbon atoms of the alkanediyl chain in 1,2, 1,3 or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, may be d to form a (C3- C6)-cycloalkyl ring or a phenyl ring, B is a bond or a group of the formula WO 87716 O R20 O ** * P ** * N ** * Q1 Q2 , O , R18 R19 O , * ** N ** H or * N R21 R22 where * marks the linkage site with L1, ** marks the linkage site with L2, P is O or NH, Q1 is a 4- to 7-membered heterocycle, Q2 is a 3- to 7-membered carbocycle or a 4- to 7-membered heterocycle, R18 is hydrogen or (C1-C4)-alkyl, R19 is hydrogen or the side group of a natural α-amino acid or of its homologues or isomers, R20 is hydrogen or (C1-C4)-alkyl, R19 and R20 er with the atoms to which they are bonded form a pyrrolidinyl ring, R21 is hydrogen or (C1-C4)-alkyl, R22 is en or (C1-C4)-alkyl, R21 and R22 together with the atoms to which they are bonded form a 3- to 7-membered carbocycle, R27 is hydrogen or (C1-C4)-alkyl, L2 is linear (C2-C10)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, where 0)-alkanediyl may be substituted by 1 to 4 substituents selected ndently of one another from the group consisting of methyl, hydroxy and benzyl, where two carbon atoms of the alkanediyl chain in 1,2, 1,3 or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, may be bridged to form a (C3-C6)-cycloalkyl ring or a phenyl ring, D is a group of the formula O #3 T1 #3 R5 N or #3 R3 R4 O , R26 T2 R1 R2 in which #3 marks the linkage site with the nitrogen atom, R1 is hydrogen or methyl, R2 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, 1-hydroxyethyl, 4-hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, yl- ethyl, ylmethyl, 1H-imidazolylmethyl or 1H-indolylmethyl, 2012/075277 R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or ic, optionally substituted heterocycle of the formula O O O or N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the yl group, R6 is hydrogen, y or benzyloxy, R3 is hydrogen or methyl, R4 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, 1-hydroxyethyl, 4-hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, 1-phenylethyl , diphenylmethyl, 1H-imidazolylmethyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9, -C(=O)-NH-NH-R10 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen or , R9 is hydrogen, methyl, ethyl, n-propyl or benzyl, R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to ered heterocycle, R10 is benzoyl, R11 is benzyl, which may be tuted in the phenyl group by methoxycarbonyl or carboxyl, R5 is hydrogen, methyl or a group of the formula WO 87716 #9 O #9 S , , #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHC(R26)-T2, R12 is phenyl which may be substituted by methoxycarbonyl, carboxyl or a group of the formula –S(O)2OH, R13 is phenyl which may be substituted by methoxycarbonyl or carboxyl, R26 is hydrogen or hydroxy, T2 is phenyl, benzyl, 1H-indolyl or 1H-indolylmethyl, R35 is methyl or hydroxy, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present ion are also compounds of the formula (XXXI) in which L1 is a bond, linear (C2-C6)-alkanediyl or a group of the formula ##1 ##2 O m where m is a number 2 or 3, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C2-C6)-alkanediyl may be substituted by 1 or 2 methyl substituents, B is a bond or a group of the formula H R20 N ** * N ** * , R18 R19 O , O O * ** or ** N * N R21 R22 R27 where * marks the linkage site with L1, ** marks the linkage site with L2, R18 is hydrogen, R19 is methyl, yl, 2-methylpropanyl or 1-methylpropanyl, R20 is hydrogen or (C1-C4)-alkyl, R19 and R20 together with the atoms to which they are bonded form a pyrrolidinyl ring, R21 is hydrogen or methyl, R22 is hydrogen or methyl, R21 and R22 together with the atoms to which they are bonded form a cyclopropyl ring, R27 is hydrogen or methyl, L2 is linear )-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ## 3 marks the e site with the group B, ##4 marks the linkage site with the nitrogen atom, where (C2-C10)-alkanediyl may be substituted by 1 or 2 methyl substituents, where two carbon atoms of the alkanediyl chain in 1,4-relation to one another, with inclusion of any carbon atoms situated n them, may be bridged to form a phenyl ring, D is a group of the formula O #3 T1 #3 R5 N or #3 R3 R4 O , R1 R2 in which #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is 1-hydroxyethyl, benzyl, 4-hydroxybenzyl, 1-phenylethyl or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the nt nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula O O O or N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the yl group, R6 is hydrogen, hydroxy or benzyloxy, R3 is hydrogen, R4 is 1-hydroxyethyl, benzyl, 4-hydroxybenzyl, 1-phenylethyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a ) phenylcyclopropan-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9, -C(=O)-NH-NH-R10 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, utyl, benzyl or tylmethyl, R8 is hydrogen or methyl, R9 is hydrogen, methyl, ethyl, n-propyl or benzyl, R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to 7-membered heterocycle, R10 is l, R11 is benzyl, which may be substituted in the phenyl group by methoxycarbonyl or carboxyl, R5 is hydrogen, methyl or a group of the formula #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHCH2phenyl, R12 is phenyl which may be substituted by methoxycarbonyl, carboxyl or a group of the formula –S(O)2OH, R13 is phenyl which may be substituted by methoxycarbonyl or carboxyl, R35 is methyl or hydroxy, and also their salts, solvates and solvates of the salts.
Particularly preferred in the context of the present ion are also compounds of the formula (XXXI) in which L1 is a bond, B is a bond, L2 is linear (C2-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, D is a group of the formula O #3 T1 N or #3 R3 R4 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is benzyl, 4-hydroxybenzyl or olylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the e site with the adjacent nitrogen atom, #5 marks the linkage site with the yl group, the ring A with the N-O moiety present therein is a mono- or ic, optionally substituted heterocycle of the formula in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy or benzyloxy, R3 is hydrogen, R4 is benzyl, 4-hydroxybenzyl or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7 or -C(=O)-NR8R9, in which 2012/075277 R7 is hydrogen, R8 is hydrogen, R9 is hydrogen, R35 is methyl, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia), in which AK is AK1 where AK1 is an antibody or an antigen-binding dy fragment which binds to FGFR2 and is bonded via the sulphur atom of a cysteine residue of the binder to the group G, G is a group of the formula N #2 where #1 marks the linkage site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, n, L1, B, L2, D and R35 have the definitions ted above, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia), in which AK is AK2 where AK2 is an antibody or an n-binding antibody fragment which binds to FGFR2 and is bonded via the NH side group of a lysine residue of the binder to the group G, G is carbonyl, n, L1, B, L2, D and R35 have the definitions indicated above, and also their salts, solvates and solvates of the salts.
Preference in the context of the present invention is also given to compounds of the formula (Ia), in which AK is AK1 where AK1 is a binder which binds FGFR2, and which is bonded via the sulphur atom of a cysteine residue of the binder to the group G, G is a group of the formula N #2 where #1 marks the linkage site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, n, L1, B, L2, D and R35 have the definitions ted above, and also their salts, solvates and solvates of the salts.
Preference in the context of the present invention is also given to nds of the formula (Ia), in which AK is AK2 where AK2 is an a binder which binds FGFR2 and which is bonded via the NH side group of a lysine residue of the binder to the group G, G is carbonyl, n, L1, B, L2, D and R35 have the definitions indicated above, and also their salts, solvates and es of the salts.
Preference in the context of the present invention is also given to compounds of the general formula (Ia), in which AK is AK2 where AK2 is a binder which binds FGFR2 and which is bonded via the NH side group of a lysine residue of the binder to the group G, G is carbonyl, L1 is a bond, B is a bond, L2 is linear (C3-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ##3 marks the e site with the group B, ##4 marks the linkage site with the nitrogen atom, n, D and R35 have the definitions indicated above, and also their salts, solvates and solvates of the salts.
Preference in the context of the present invention is also given to compounds of the general formula (Ia), in which AK is AK1 where AK1 is a binder which binds FGFR2 and which is attached via the sulphur atom of a cysteine residue of the binder to the group G, G is a group of the formula N #2 where #1 marks the e site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, L1 is a bond, linear )-alkanediyl or a group of the formula ##1 ##2 O m where m is a number 2 or 3, ##1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C3-C5)-alkanediyl may be substituted by 1 or 2 methyl substituents, B is a bond or a group of the formula O O L3 ** * N N L4 R16 R17 where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a bond or a group of the formula O **** R25 O in which *** marks the linkage site with the yl group, **** marks the linkage site with L2, R25 is methyl, R28 is hydrogen, methylcarbonyl or tert-butyloxycarbonyl, R16 is hydrogen or methyl, R17 is hydrogen or methyl, or R16 and R17 together with the atoms to which they are bonded form a zinyl ring, L2 is linear (C3-C5)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ##3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, n, D and R35 have the definitions indicated above, and also their salts, solvates and solvates of the salts.
Preferred in the t of the present invention are also compounds of the formula (Ia), (XXXa) and (XXXI), in which L1 is a bond, B is a bond, L2 is linear (C3-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ##3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, n, AK, Cys, G, D and R35 have the definitions indicated above, and also their salts, solvates and solvates of the salts.
Preferred in the t of the present invention are also compounds of the formula (Ia), in which L1 is linear 0)-alkanediyl or a group of the formula ##1 ##2 O m where m is a number from 2 to 6, ##1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C1-C10)-alkanediyl may be substituted by 1 to 4 substituents ed ndently of one another from the group consisting of methyl, hydroxyl and benzyl, B is a bond or a group of the formula O O O L3 ** O ** * N N L4 * Q1 R16 R17 O , , O R36 O ** * N or O ** H * R23 R24 O R37 O where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or (C2-C4)-alkanediyl, L4 is a group of the formula R28 HN HN O O **** or *** **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is hydrogen or methyl, R28 is hydrogen, (C1-C4)-alkylcarbonyl, utyloxycarbonyl or benzyloxycarbonyl, Q1 is a 4- to 7-membered cycle, R16 is hydrogen or (C1-C4)-alkyl, R17 is hydrogen or (C1-C4)-alkyl, R16 and R17 together with the atoms to which they are bonded form a 5- or 6-membered heterocycle, R23 is (C1-C4)-alkyl, R24 is hydrogen or (C1-C4)-alkyl, R36 is hydrogen, (C1-C4)-alkylcarbonyl, utyloxycarbonyl or benzyloxycarbonyl, R37 is hydrogen or methyl, R36 and R37 together with the atoms to which they are bonded form a pyrrolidine ring, L2 is linear (C2-C10)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, ##3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, where (C2-C10)-alkanediyl may be substituted by 1 to 4 tuents selected independently of one another from the group consisting of methyl, hydroxyl and benzyl, n, AK, G, D and R35 have the tions indicated above, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia), in which L1 is linear (C2-C6)-alkanediyl or a group of the formula ##1 ##2 O m where m is a number 2 or 3, ##1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, B is a bond or a group of the formula O O R36 L3 ** * N N L4 or O ** R16 R17 R37 O where * marks the e site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a group of the formula R28 HN HN O O **** or *** **** R25 O where *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is hydrogen or methyl, R28 is hydrogen, carbonyl or utyloxycarbonyl, R16 is hydrogen or methyl, R17 is hydrogen or methyl, R16 and R17 together with the atoms to which they are bonded form a piperazinyl ring, R36 is hydrogen, methylcarbonyl or tert-butyloxycarbonyl, R37 is hydrogen or methyl, R36 and R37 together with the atoms to which they are bonded form a pyrrolidine ring, L2 is linear (C2-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ##3 marks the linkage site with the group B, ##4 marks the linkage site with the en atom, n, AK, G, D and R35 have the definitions indicated above, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia) and (XXXa), in which G is a group of the a N #2 in which #1 marks the linkage site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, L1 is linear (C3-C5)-alkanediyl or a group of the formula ##1 ##2 O m in which m is a number 2 or 3, ##1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C3-C5)-alkanediyl may be substituted by 1 or 2 methyl substituents, B is a bond or a group of the formula O O L3 ** * N N L4 R16 R17 where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a bond, L2 is linear (C3-C5)-alkanediyl or is a group of the a ##3 ##4 O p where p is a number 2 or 3, ##3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, n, AK1, Cys, D, R16 and R17 have the definitions ted above, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia), (XXXa) and (XXXI), in which D is a group of the formula O #3 R5 #3 T1 N or #3 R3 R4 O , R26 T2 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen or methyl, R2 is pyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, 1-hydroxyethyl, 4-hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, 1-phenyl- ethyl, diphenylmethyl, 1H-imidazolylmethyl or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent en atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula O O or O N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy or oxy, R3 is hydrogen or , R4 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, 1-hydroxyethyl, 4- hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, 1-phenylethyl , diphenylmethyl, 1H-imidazolylmethyl or 1H-indolylmethyl, R3 and R4 er with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9, -NH-NH-R10 or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl or adamantylmethyl, R8 is hydrogen or , R9 is hydrogen, methyl, ethyl, n-propyl or benzyl, R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to 7- membered heterocycle, R10 is benzoyl, R11 is benzyl, which may be substituted in the phenyl group by ycarbonyl or carboxyl R5 is hydrogen, methyl or a group of the formula #9 O #9 S , , #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with 26)-T2, R12 is phenyl which may be substituted by methoxycarbonyl, yl or a group of the formula –S(O)2OH, R13 is phenyl which may be tuted by methoxycarbonyl or carboxyl, R26 is hydrogen, T2 is phenyl, benzyl, 1H-indolyl or 1H-indolylmethyl, n, AK, Cys, G, L1, B, L2, D and R35 have the definitions indicated above, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia), (XXXa) and (XXXI), in which D is a group of the formula R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen or methyl, R2 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, 1-hydroxyethyl, 4- hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxyaminobenzyl, 1- ethyl, diphenylmethyl, dazolylmethyl or 1H-indolylmethyl, or R1 and R2 together with the carbon atom to which they are bonded form a ) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present n is a mono- or bicyclic, optionally substituted heterocycle of the formula O O O or N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy or benzyloxy, n, AK, Cys, G, L1, B, L2 and R35 have the definitions indicated above, and also their salts, solvates and es of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia), (XXXa) and (XXXI), in which D is a group of the formula R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is , 4-hydroxybenzyl, 1-phenylethyl or 1H-indolylmethyl, or R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R) phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the nt nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula WO 87716 in which #6 marks the linkage site with the carbonyl group, n, AK, Cys, G, L1, B, L2 and R35 have the definitions indicated above, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia), (XXXa) and , in which R35 is hydroxyl, and n, AK, Cys, G, L1, B, L2, D and R35 have the tions indicated above, and also their salts, solvates and solvates of the salts.
Preferred in the context of the present invention are also compounds of the formula (Ia), (XXXa) and (XXXI), in which R35 is methyl, n, AK, Cys, G, L1, B, L2, D and R35 have the definitions indicated above, and also their salts, es and solvates of the salts.
A preferred subject of the present invention are binder-drug conjugates of the general formula (Ia) in which D can have the following structures and * stands for the linkage site with the nitrogen atom: or , and also their salts, solvates and solvates of the salts.
A preferred t of the present invention are binder-drug conjugates of the l formula (Ia) in which D has a structure which is disclosed by one of the intermediates of the present invention; and the linker unit -B-L2-§§ and also all other variables are defined in accordance with the present invention; and their salts, solvates and solvates of the salts. AK is preferably an anti- FGFR2 antibody or antigen-binding fragment thereof.
A red subject of the present invention are binder-drug conjugates of the general formula (Ia) in which the linker-drug unit has a structure which is disclosed by one of the intermediates or es of the present invention; and their salts, solvates and solvates of the salts. AK is preferably an anti-FGFR2 antibody or antigen-binding fragment thereof.
A preferred subject of the present invention are binder-drug conjugates of the general formula (Ia) in which the linker-drug unit has a ure which is disclosed by one of the examples of the present invention; and their salts, es and solvates of the salts. AK is preferably an anti- FGFR2 antibody or antigen-binding fragment thereof.
Particularly red subject of the present invention are binder-drug conjugates of the general a (Ia) H C R35 H C 3 CH 3 O 3 H N N N D AK G L1 B L2 N N O O CH O CH3 O O CH H C CH CH3 3 3 3 (Ia), in which n is a number from 1 to 50, AK is a binder which binds to FGFR2, the group §-G-L1-B-§§ is a linker, where § marks the linkage site with the group AK and §§ marks the linkage site with the nitrogen atom, L2 is linear (C2-C10)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, WO 87716 ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, where 0)-alkanediyl may be tuted by 1 to 4 methyl substituents ed independently of one another from the group consisting of methyl, hydroxyl and benzyl and where two carbon atoms of the alkanediyl chain in 1,2, 1,3 or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, may be bridged to form a (C3-C6)-cycloalkyl ring or a phenyl ring, D is a group of the following formula, where * is the linkage site to the nitrogen atom H and also their salts, solvates and solvates of the salts.
Particularly preferred subject of the present invention are compounds of the following formula H C CH H C 3 3 3 CH3 N O H H N AK N N O N N O O O CH3 O CH O O 3 CH3 H C CH CH 3 3 3 where AK is a binder which binds FGFR2, and n is a number from 1 to 10, and also their salts, solvates and solvates of the salts. It is preferred if the binder is bonded via a NH side group of a lysine residue to the linker-toxophore unit.
Particularly preferred subject of the present invention are compounds of the following formula H C CH H C 3 3 3 CH3 N O H H N AK N N O N N O O O CH3 O CH O O 3 CH3 H C CH CH 3 3 3 where AK is an antibody or an antibody fragment which binds FGFR2, and n is a number from 1 to 10, and also their salts, solvates and solvates of the salts. It is preferred if the antibody or antibody fragment is bonded via an NH side group of a lysine residue of the antibody or antibody fragment to the linker-toxophore unit.
Particularly preferred subject of the present invention is the compound of the following a H C CH H C 3 3 3 CH 3 N O H H N AK N 2A N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 where AK2A is M048-D01-hIgG1 and n is a number from 1 to 10, and also the salts, solvates and solvates of the salts thereof.
A further ularly preferred subject of the present ion is the compound of the following formula H3C CH H3C 3 CH O 3 N H N AK2B N N O N N O O O CH O CH3 O O 3 CH H C CH3 CH 3 3 3 where AK2B is M048-D01-hIgG1-b and n is a number from 1 to 10, and also the salts, solvates and solvates of the salts f.
The definitions of radicals that are indicated individually in the respective combinations and preferred combinations of radicals are also replaced arbitrarily by radical tions of other combinations, independently of the respective combinations of radicals that are indicated.
Especially preferred are combinations of two or more of the abovementioned preference ranges.
Further ed by the invention is a s for preparing the compounds of the invention of the formula (Ia), characterized in that a solution of the binder in PBS buffer [A] is admixed with a suitable reducing agent, such as, for example, dithiothreitol or tris(2- carboxyethyl)phosphine hydrochloride, and is uently reacted with a compound of the formula (IIa) O H C R35 H C 3 CH 3 O 3 H N N N D N L1 B L2 N N O O CH3 O CH O O 3 CH H C CH CH 3 O 3 (IIa), 3 3 in which D, L1, B, L2 and R35 each have the definitions indicated above, to give a compound of the formula (I-A) O H C R35 H C 3 CH 3 O 3 H AK1 H N N N D N L1 B L2 N N O O CH O CH O O 3 CH 3 CH 3 O H C CH 3 3 3 (Ia-A), in which n, AK1, D, L1, B, L2 and R35 each have the tions indicated above, or [B] is reacted with a compound of formula (IIIa) H3C R35 H3C O CH3 O H O H N N N D N L1 B L2 N N O O CH O CH O O CH O 3 3 3 H C CH CH 3 3 (IIIa), in which D, L1, B, L2 and R35 each have the definitions indicated above, to give a compound of the formula (Ia-B) O H C R35 H C 3 CH 3 O 3 H AK H N 2 N N D L1 B L2 N N O O CH O CH O O 3 3 CH CH 3 H C CH 3 3 3 (Ia-B), in which n, AK2, D, L1, B, L2 and R35 each have the definitions indicated above.
Cysteine coupling: The partial reduction of the antibody and also the subsequent conjugation of the (partially) reduced antibody with a compound of the a (IIa) takes place in accordance with the methods known to the skilled person, see e.g. Ducry et. al., Bioconj. Chem. 2010, 21, 5 and references herein, Klussman et. al., Bioconj. Chem. 2004, 15(4), 765-773. The mild reduction of the antibody is accomplished preferably by addition of 2-6 equivalents of TCEP to the antibody, which is present in a suitable buffer solution, preferably phosphate buffer, and by stirring for 30-180 minutes at temperatures between 15 and 40°C, preferably at RT. This is ed by the ation, by addition of a solution of a compound of the formula (IIa) in DMSO, acetonitrile or DMF to the solution of the (partially) reduced antibody in PBS buffer, and subsequent on at a ature of 0°C to +40°C, more particularly of +10°C to +30°C, for a period of 30 minutes to 6 hours, more particularly 1 to 2 hours.
Lysine ng: First of all the nds of the formula (IIIa) or comparable activated carboxyl components are ed by conventional methods of peptide chemistry. They are then taken up in inert solvents such as DMSO or DMF, for example, and added to the antibody, which is preferably present in phosphate buffer at a neutral pH. The solution is stirred for 1-16 h at a temperature between 15 and 40°C, preferably RT.
The preparation processes described above are elucidated by way of e using the schemes below (Scheme 1 and 2): Scheme 1 H C CH H C O 3 3 N O 3 CH 3 H H H N N N N O N N N H O O O N O CH O CH O O 3 O 3 3 H C CH 3 3 CH O 3 1 H C CH H C O 3 3 O 3 CH N 3 H H H N N N N N O N N N H O O O O CH O CH O O CH 3 C CH 3 3 3 3 CH [a): 1. AK (antibody), TCEP, PBS buffer, RT; 2. Addition of the ide tive in DMSO, RT].
Scheme 2 H C CH H C 3 3 CH NH O O 3 3 H 2 H N O N N O N N N O O O CH3 O CH O O CH 3 3 O H3C CH 3 CH3 H C CH3 H C 3 3 CH NH O 3 2 AK H N 2 N N O N N O O O CH O CH3 O O CH3 3 H3C CH 3 CH3 [a): AK (antibody), PBS buffer, RT admix with activated carboxyl derivative of the linker-drug components].
The compounds of the formula (II) in which L1 and B are a bond can be prepared by subjecting a compound of the formula (IV) H C CH H C 3 3 CH O 3 H N N N D HN N O O CH O CH O O 3 CH 3 CH 3 H C CH 3 (IV), 3 3 in which D has the definition indicated above, to reductive amination in an inert solvent with a compound of the formula (V) PG1 N L2A H (V), in which L2A has the above-defined definition of L2, but is shortened by one carbon atom in the alkyl chain length, PG1 is an amino-protective group such as, for example, (9H-fluorenylmethoxy)carbonyl, tert-butoxycarbonyl or benzyloxycarbonyl, to give a compound of the formula (VI) H C CH H C 3 3 CH O 3 H N PG1 L2 N N D N N N H O O CH O CH O O CH 3 3 3 H C CH CH 3 (VI), 3 3 in which D, L2 and PG1 have the tion indicated above, eliminating the tive group PG1 from this compound by methods known to the skilled , and reacting the deprotected compound in an inert solvent in the presence of a suitable base with methyl 2,5-dioxo-2,5-dihydro-1H-pyrrolecarboxylate to give a compound of the formula (II-A) H C CH3 H C 3 CH 3 3 O O H H N L2 N N D N N N O O CH O CH O O 3 3 CH O H C CH CH3 (II-A), 3 3 in which D and L2 each have the definitions indicated above.
The compounds of the formula (II) in which B is a group of the formula (B1) * ** N N R14 R15 (B1), in which *, **, R14 and R15 each have the conditions indicated above, can be prepared by eliminating the protective group PG1 from a compound of the formula (VI) by methods known to the skilled person, and reacting the deprotected compound in an inert solvent in the presence of a suitable base with a compound of the formula (VII) O O N L1 N O (VII), in which L1 has the definition indicated above, to give a compound of the formula (II-B) H C CH3 H C 3 CH 3 3 O O O H H N L1 L2 N N D N N N N N H H O O CH O CH O O CH 3 3 3 O H C CH CH3 (II-B), 3 3 in which D, L1 and L2 each have the tions indicated above.
The compounds of the formula (II) in which B is a group of the formula (B2) O O * N N ** R16 R17 (B2), in which *, **, L3, R16 and R17 each have the ions indicated above can be prepared by subjecting a compound of the formula (IV) to reductive ion in an inert solvent with a compound of the a (VIII) O O HO L2A H (VIII), in which L2A has the above-defined definition of L2, but is ned by one carbon atom in the alkyl chain length, to give a compound of the formula (IX) H C CH H C 3 CH 3 3 O 3 H H N HO L2 N N D N N O O O CH O CH O O CH 3 3 H C CH CH 3 3 (IX), 3 3 in which D and L2 have the tions indicated above, and reacting this compound in an inert solvent in the presence of a suitable coupling reagent and a suitable base with a compound of the formula (X) N L1 N L3 NH H 2 O (X), in which L1 and L3 each have the definitions indicated above, to give a compound of the formula (II-C) H C CH3 H C 3 CH 3 3 O O H H H H N L1 N N L2 N N D N L3 N N O O O O CH O CH O O 3 3 CH O H C CH CH3 3 3 (II-C), in which D, L1, L2 and L3 each have the tions indicated above.
Compound of the formula (II), in which B is a group of the formula (B3) O O L3 ** * N N L4A R16 R17 (B3), in which *, **, L3, R16 and R17 each have the conditions indicated above and L4A is a group of the formula 2 O O **** or *** **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is hydrogen or methyl, can be ed by reacting a compound of the formula (IX) in an inert solvent in the presence of a suitable base and a suitable coupling t with a compound of the formula (XI-A) or (XI-B) O R25 O PG2 PG2 O OH O HN N PG1 (XI-A) or PG1 (XI-B), in which R25 and PG1 each have the definitions ted above and PG2 is a suitable carboxyl-protective group, more particularly benzyl, to give a compound (XII-A) or (XII-B) 2012/075277 PG1 H3C CH H3C CH3 HN 3 O H H N O O L2 N N D PG2 N N O O O R25 O CH O CH O O CH 3 3 3 H C CH CH 3 3 (XII-A) H C CH H C 3 3 CH O 3 H H N O L2 N N D N N PG1 N O O O CH O CH3 O O CH H CH CH 3 C 3 3 3 ), in which D, PG1, PG2 and L2 have the definitions indicated above, eliminating the protective group PG2 from this compound subsequently, by methods known to the skilled person, and reacting the deprotected compound in an inert solvent in the presence of a suitable coupling reagent and a suitable base with a compound of the formula (X), and finally, ating the protective group PG1 from this compound, by methods known to the skilled person, to give a compound of the formula (II-D-A) or (II-D-B) H C CH H C NH 3 3 CH O 3 2 H H H N N O L2 N N D L3 N N O O O NH O R25 O CH O CH O O CH 3 3 3 H C CH CH O 3 3 (II-D-A), 2012/075277 H C CH H C 3 3 CH O 3 H N O L2 N N D N N HN O O O CH O CH O O CH 3 3 3 H C CH CH 3 3 O O L3 L1 N O O (II-D-B), in which D, L1, L2 and L3 have the definitions indicated above.
Compound of the formula (II), in which B is a group of the formula (B4) O ** * Q1A O (B4), in which *, ** each have the conditions indicated above and Q1A is an N-linked 4- to 7-membered heterocycle, can be prepared by reacting a compound of the a (IX) in an inert solvent in the presence of a suitable base and a suitable coupling reagent with a compound of the formula (XXI) PG1 (XXI), in which PG1 and Q1A each have the definitions indicated above, to give a compound of the formula (XXII) H C CH H C 3 CH 3 O 3 H N O L2 N N D N N N O O PG1 O CH O CH O O CH 3 3 3 H C CH CH 3 3 (XXII), in which PG1, Q1A, D and L2 have the definitions indicated above, ating the protective group PG1 from this compound, by methods known to the skilled person, and subsequently reacting the deprotected compound in an inert solvent in the presence of a suitable coupling reagent and a suitable base with a compound of the formula (XXIII) N L1 O (XXIII), in which L1 has the definition ted above, to give a compound of the formula (II-D) H3C CH H3C 3 CH3 O H H N O O L2 N N D N N L1 N Q1A O O N O CH3 O CH O O 3 CH H C CH CH 3 O 3 3 O (II-D), in which Q1A, D, L1 and L2 have the definitions indicated above.
The compounds of the formula (III), in which L1 and B are a bond can be prepared by reacting a compound of the a (IX) in an inert solvent in the presence of a suitable coupling t and a suitable base with N-hydroxysuccinimide to give a compound of the formula (III-A) H C CH3 H C 3 CH 3 3 O O H H N O L2 N N D N N N O O O CH O CH O O CH 3 3 3 O H C CH CH3 (III-A), 3 3 in which D and L2 each have the tions indicated above.
The compounds of the formula (III), in which L1 is a bond and B is a group of the a (B5A) * P ** O (B5A), in which *, ** and P each have the definitions indicated above and Q2A is a 3- to 7-membered carbocycle, can be prepared by reacting a compound of the formula (IX) in an inert solvent in the ce of a suitable coupling reagent and a suitable base with a compound of the formula (XIII) PG2 P H O Q2A (XIII), in which P, Q2A and PG2 each have the definitions indicated above, to give a compound of the formula (XIV) H C CH3 H C 3 CH O 3 O 3 H H N PG2 P L2 N N D O Q2A N N O O O CH O CH O O 3 3 CH H C CH CH3 3 (XIV), 3 3 in which D, P, Q2A, L2 and PG2 each have the definitions indicated above, eliminating the protective group PG2 from this compound by methods known to the skilled person, and subsequently reacting the deprotected nd in an inert solvent in the presence of a suitable base with N-hydroxysuccinimide to give a compound of the formula (III-B) O H C CH3 H C 3 CH O 3 O 3 H H N N P L2 N N D O Q2A N N O O O O CH O CH O O 3 3 CH CH3 3 H C CH 3 3 (III-B), in which D, P, Q2A and L2 each have the tions indicated above.
The compounds of the formula (III), in which L1 is a bond and B is a group of the formula (B6) * N ** R18 R19 O (B6), in which *, **, R18, R19 and R20 each have the definitions indicated above, can be prepared by reacting a compound of the formula (IX) in an inert solvent in the presence of a suitable coupling reagent and a suitable base with a nd of the formula (XV) O R20 PG2 NH R18 R19 (XV), in which R18, R19, R20 and PG2 each have the tions indicated above, to give a compound of the formula (XVI) H C CH3 H C 3 CH O R20 3 O 3 H H N PG2 N L2 N N D O N N O O R18 R19 O CH O CH O O 3 CH 3 CH3 3 H C CH (XVI), 3 3 in which D, R18, R19, R20, L2 and PG2 each have the definitions indicated above, eliminating the protective group PG2 from this compound by s known to the skilled person, and subsequently reacting the deprotected compound in an inert solvent in the presence of a suitable coupling reagent and a suitable base with N-hydroxysuccinimide to give a compound of the formula (III-C) O H C CH3 H C 3 CH O R20 3 O 3 H H N N N L2 N N D O N N O O O R18 R19 O CH O CH O O CH 3 3 H C CH CH3 3 3 3 (III-C), in which D, R18, R19, R20 and L2 each have the definitions indicated above.
The nds of the formula (III), in which L1 is a bond and B is a group of the formula (B7) * ** R21 R22 (B7), in which *, **, R21 and R22 each have the definitions indicated above, can be prepared by eliminating the protective group PG1 from a compound of the formula (VI) by methods known to the skilled person, and reacting the resultant deprotected compound in an inert solvent in the ce of a le base with a compound of the formula (XVII) O R21 R22 O O O N N O O O O (XVII), in which R21 and R22 each have the definitions ted above, to give a compound of the formula (III-D) O H C CH3 H C 3 CH O O 3 O 3 H H N N L2 N N D O N N N H O O O R21 R22 CH O CH O O 3 CH 3 CH3 3 H C CH 3 3 (III-D), in which D, R21, R22 and L2 each have the definitions indicated above.
The compounds of the formula (III), in which B is a group of the formula (B8) O ** * N R23 R24 O (B8), in which *, **, R23 and R24 each have the definitions indicated above, can be prepared by ng a compound of the a (IX) in an inert solvent in the presence of a suitable coupling reagent and a suitable base with a compound of the formula (XVIII) PG1 OH R23 R24 (XVIII), in which R23, R24 and PG1 each have the definitions indicated above, to give a compound of the formula (XIX) H C CH3 H C 3 CH 3 O 3 H H N PG1 O L2 N N D N N N H O O R23 R24 O CH O CH O O 3 CH 3 3 H C CH CH3 (XIX), 3 3 in which D, R23, R24, L2 and PG1 each have the definitions indicated above, ating the protective group PG1 from this compound by methods known to the skilled person, and subsequently reacting the deprotected compound in an inert solvent in the presence of a suitable coupling reagent and a suitable base with a compound of the formula (XX) O O O L1A O N N O O O O (XX), in which L1A is linear (C1-C10)-alkanediyl or is a group of the formula ##1 ##2 O m where m is a number from 2 to 6, ##1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C1-C10)-alkanediyl may be substituted by 1 to 4 methyl substituents, and where two carbon atoms of the diyl chain in 1,2, 1,3 or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, may be bridged to form a (C3- cloalkyl ring or a phenyl ring, to give a compound of the formula (III-E) H C CH H C 3 CH O 3 O 3 H H N O L2 N N D L1A N N N H O O R23 R24 O CH O CH3 O O CH3 O O 3 H C CH CH 3 3 O N (III-E), in which D, R23, R24, L1A and L2 each have the definitions indicated above.
The compounds of the a (III), in which B is a group of the formula (B5B) * O ** O (B5B), in which * and ** each have the definitions indicated above and Q2B is an N-linked 4- to ered heterocycle, can be prepared by ng a compound of the formula (IX) in an inert solvent in the presence of a suitable base and a suitable coupling reagent with a compound of the formula (XXIV) PG1 (XXIV), in which PG1 and Q2B each have the definitions indicated above, to give a compound of the formula (XXV) H C CH H C 3 CH 3 3 O 3 H H N O L2 N N D N N N O O PG1 O CH O CH O O CH3 3 3 H3C CH3 CH (XXV), in which PG1, Q2B, D and L2 have the definitions indicated above, eliminating the protective group PG1 from this compound by methods known to the skilled person, and subsequently converting the deprotected compound in an inert solvent in the presence of a suitable base with a compound of the formula (XX) into a compound of the formula (III-F) H C CH H C 3 CH 3 3 O 3 H N O L2 N N D N N O N Q2B O O O CH O CH O O 3 CH 3 3 H C CH CH O L1A 3 3 (III-F), in which Q2B, D, L1A and L2 have the definitions ted above.
The reactions (IV) + (V) → (VI) and (IV) + (VIII) → (IX) take place in the solvents which are customary for a reductive amination and are inert under the reaction conditions, optionally in the presence of an acid and/or of a water-removing agent as catalyst. Such solvents include, for e, alcohols such as methanol, l, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis(2-methoxyethyl) ether, or other solvents such as dichloromethane, 1,2-dichloroethane, N,N-dimethylformamide or else water.
It is also possible to use mixtures of these ts. As solvent it is preferred to use a 1,4-dioxane/water mixture, with on of acetic acid or dilute hydrochloric acid as catalyst. ng agents suitable for this reaction are, in particular, complex borohydrides, such as, for example, sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, tetra- n-butylammonium borohydride or borane-pyridine complex. It is preferred to use sodium cyanoborohydride or borane-pyridine complex.
The reactions (IV) + (V) → (VI) and (IV) + (VIII) → (IX) take place in l in a temperature range from 0°C to +120°C, preferably at +50°C to +100°C. The reactions may be carried out under atmospheric, increased or d pressure (e.g. from 0.5 to 5 bar); it is usual to operate at atmospheric pressure.
The above-described coupling reactions (IX) + (X) → (II-C), (XII-A) or (XII-B) + (X) → (II-D-A) or (II-D-B), (IX) + (XIII) → (XIV), (IX) + (XV) → (XVI) and (XXII) + (XXIII) → (II-D) (amide formation from amine component and carboxylic acid component tively) are carried out by standard methods of peptide chemistry [see e.g. M. Bodanszky, Principles of Peptide Synthesis, Springer-Verlag, Berlin, 1993; M. Bodanszky and A. Bodanszky, The Practice of Peptide Synthesis , Springer-Verlag, Berlin, 1984; H.-D. Jakubke and H. eit, äuren, Peptide, Proteine , Verlag Chemie, Weinheim, 1982].
Examples of inert solvents for these coupling reactions are ethers such as diethyl ether, diisopropyl ether, tert -butyl methyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis(2- methoxyethyl) ether, arbons such as benzene, toluene, , pentane, hexane, heptane, cyclohexane or petroleum ons, halogenated hydrocarbons such as romethane, trichloromethane, tetrachloromethane, 1,2-dichloroethane, trichloroethylene or chlorobenzene, or dipolar-aprotic solvents such as acetone, methyl ethyl ketone, acetonitrile, ethyl acetate, pyridine, dimethyl sulphoxide (DMSO), N,N -dimethylformamide (DMF), N,N -dimethylacetamide (DMA), N,N' -dimethylpropyleneurea (DMPU) or N-methylpyrrolidinone (NMP). It is also le to use mixtures of such solvents. ence is given to using N,N -dimethylformamide.
Examples of suitable activating/condensing agents for these couplings include carbodiimides such as N,N' -diethyl-, N,N' -dipropyl-, N,N' -diisopropyl-, N,N' -dicyclohexylcarbodiimide (DCC) or N-(3- dimethylaminoisopropyl)-N' -ethylcarbodiimide hydrochloride (EDC), phosgene derivatives such as N,N' -carbonyldiimidazole (CDI) or isobutyl chloroformate, 1,2-oxazolium compounds such as 2-ethylphenyl-1,2-oxazolium 3-sulphate or 2-tert -butylmethylisoxazolium perchlorate, acylamino compounds such as 2-ethoxyethoxycarbonyl-1,2-dihydroquinoline, phosphorus compounds such as propanephosphonic ide, diethyl cyanophosphonate, bis(2-oxo oxazolidinyl)phosphoryl chloride, benzotriazolyloxytris(dimethylamino)phosphonium hexafluorophosphate or benzotriazolyloxytris(pyrrolidino)phosphonium hexafluorophosphate (PyBOP), or uronium compounds such as O-(benzotriazolyl)-N,N,N',N' -tetramethyluronium luoroborate (TBTU), O-(benzotriazolyl)-N,N,N',N' -tetramethyluronium hexafluorophosphate (HBTU), xo(2H)-pyridyl)-1,1,3,3-tetramethyluronium tetrafluoroborate (TPTU), O-(7-azabenzotriazolyl)-N,N,N',N' -tetramethyluronium hexafluorophosphate (HATU) or O-(1 Hchlorobenzotriazolyl)-1,1,3,3-tetramethyluronium- tetrafluoroborate (TCTU), optionally in ation with further auxiliaries such as 1- hydroxybenzotriazole (HOBt) or N-hydroxysuccinimide (HOSu), and also, as bases, alkali metal carbonates, e.g. sodium or potassium carbonate, or tertiary amine bases such as triethylamine, N-methylmorpholine, N-methylpiperidine, N,N -diisopropylethylamine, pyridine or 4-N,N - dimethylaminopyridine.
In the context of the present invention, as activating/condensing agents for such coupling reactions, it is preferred to use N-(3-dimethylaminoisopropyl)-N' -ethylcarbodiimide hydrochloride (EDC) in combination with 1-hydroxybenzotriazole (HOBt) and N,N-diisopropylethylamine, or O-(7-azabenzotriazolyl)-N,N,N',N' -tetramethyluronium uorophosphate (HATU) likewise in conjunction with N,N -diisopropylethylamine.
The coupling ons (IX) + (X) → (II-C), ) or (XII-B) + (X) → (II-D-A) or B), (IX) + (XIII) → (XIV) , (IX) + (XV) → (XVI) and (XXII) + (XXIII) → (II-D) are carried out in general in a temperature range from -20°C to +60°C, preferably at 0°C to +40°C. The reactions may take place under heric, at increased or at reduced pressure (e.g. from 0.5 to 5 bar); it is usual to operate under heric pressure.
The esterifications (IX) + (XVIII) → (XII) and (IX) + (XI-A) or (XI-B) → (XII-A) or (XII-B), (IX) + (XXIV) → (XXV) and also (IX) + (XXI) → (XXII) take place in analogy to the above-described amide coupling reactions. These reactions take place preferably in dichloromethane, using N-(3- dimethylaminoisopropyl)-N' -ethylcarbodiimide hydrochloride (EDC) and 4-dimethylamino- pyridine at a temperature of +50°C to 100°C under atmospheric re.
The functional groups optionally present in the compounds – such as amino, hydroxyl and yl groups in particular – may also be present in a temporarily protected form during the above-described process steps, if useful or ary. In these cases, such protective groups are introduced and removed in accordance with customary methods known from peptide chemistry [see, for example, T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, Wiley, New York, 1999; M. Bodanszky and A. zky, The Practice of Peptide Synthesis, Springer- Verlag, Berlin, 1984]. Where two or more protected groups are present, they can be liberated again optionally simultaneously in a one-pot reaction, or else liberated again in separate reaction steps.
As an amino-protective group PG1 it is preferred to use tert -butoxycarbonyl (Boc), benzyloxycarbonyl (Z) or (9H-fluorenylmethoxy)carbonyl (Fmoc); for a hydroxyl or carboxyl on it is preferred to use tert -butyl or benzyl as protective group PG2. The elimination of a tert -butyl or tert -butoxycarbonyl group is typically accomplished by treatment with a strong acid, such as hydrogen chloride, hydrogen bromide or trifluoroacetic acid, in an inert solvent such as diethyl ether, oxane, dichloromethane or acetic acid; this reaction may optionally also be carried out without addition of an inert solvent. In the case of benzyl or benzyloxycarbonyl as protective group, this group is removed preferably by hydrogenolysis in the presence of a suitable ium catalyst, such as palladium on activated carbon, for e. The (9H-fluoren oxy)carbonyl group is generally eliminated using a secondary amine base such as diethylamine or dine.
The reaction (VI) → (II-A) takes place in a t which is inert under the reaction conditions, such as, for example, ethers such as tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis(2- methoxyethyl) ether, alcohols such as methanol, ethanol, isopropanol, n-butanol or tert -butanol, or dipolar-aprotic solvents such as acetone, methyl ethyl ketone, acetonitrile, ethyl acetate, pyridine, dimethyl sulphoxide (DMSO), N,N -dimethylformamide (DMF), N,N -dimethylacetamide (DMA), N,N' -dimethylpropyleneurea (DMPU) or N-methylpyrrolidinone (NMP) or water. It is also possible to use mixtures of such ts. Preference is given to using a e of 1,4-dioxane and water.
Suitable bases for the reaction (VI) → (II-A) are, for example, alkali metal ates such as potassium carbonate, sodium carbonate or lithium carbonate, alkali metal hydrogencarbonates such as sodium or potassium hydrogencarbonate or alkali metal alkoxides such as sodium methoxide, sodium ethoxide or potassium tert-butoxide. It is preferred to use sodium hydrogencarbonate.
The reaction (VI) → (II-A) takes place in a temperature range from 0°C to +50°C, preferably at +10°C to +30°C. The reaction may take place under atmospheric, under elevated or under reduced pressure (e.g. from 0.5 to 5 bar); it is usual to operate under atmospheric pressure.
The reaction (VI) + (VII) → (II-B) takes place in a solvent which is inert under the reaction conditions, such as, for e, ethers such as ydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis(2-methoxyethyl) ether, alcohols such as methanol, ethanol, isopropanol, n-butanol or tert - butanol, or r-aprotic solvents such as acetone, methyl ethyl ketone, acetonitrile, ethyl acetate, pyridine, dimethyl sulphoxide (DMSO), N,N -dimethylformamide (DMF), N,N - dimethylacetamide (DMA), N,N' -dimethylpropyleneurea (DMPU) or N-methylpyrrolidinone (NMP) or water. It is also possible to use es of such solvents. Preference is given to using DMF.
Suitable bases for the on (VI) + (VII) → (II-B) are, for example, ry amine bases such as triethylamine, N-methylmorpholine, N-methylpiperidine, N,N -diisopropylethylamine, pyridine or 4-N,N -dimethylaminopyridine. Preference is given to using N,N -diisopropylethylamine.
The reaction (VI) + (VII) → (II-B) takes place in a temperature range from 0°C to +50°C, preferably at +10°C to +30°C. The reaction may take place under atmospheric, under elevated or under reduced pressure (e.g. from 0.5 to 5 bar); it is usual to operate under atmospheric re.
The reactions (IX) → (III-A), (XIV) → (III-B) and (XVI) → (III-C) and also (VI) + (XVII) → (III- D), (XIX) + (XX) → (III-E) and (XXV) + (XX) → (III-F) take place in a solvent which is inert under the reaction conditions. Examples of suitable ts are ethers such as diethyl ether, diisopropyl ether, tert -butyl methyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis(2-methoxyethyl) ether, hydrocarbons such as benzene, toluene, xylene, pentane, hexane, heptane, cyclohexane or petroleum ons, halogenated hydrocarbons such as dichloromethane, trichloromethane, tetrachloromethane, 1,2-dichloroethane, trichloroethylene or chlorobenzene, or dipolar-aprotic solvents such as acetone, methyl ethyl ketone, acetonitrile, ethyl acetate, pyridine, dimethyl sulphoxide (DMSO), N,N -dimethylformamide (DMF), N,N -dimethylacetamide (DMA), 2012/075277 N,N' hylpropyleneurea (DMPU) or ylpyrrolidinone (NMP). It is also possible to use mixtures of such solvents. Preference is given to using N,N hylformamide.
Suitable bases for these reactions are, for example, tertiary amines such as triethylamine, N-methylmorpholine, N-methylpiperidine, N,N propylethylamine, pyridine or 4-N,N - ylaminopyridine. Preference is given to using N,N -diisopropylethylamine, optionally with addition of 4-N,N -dimethylaminopyridine.
The reactions (IX) → (III-A), (XIV) → (III-B) and (XVI) → (III-C) and also (VI) + (XVII) → (IIID ) and (XIX) + (XX) → (III-E) take place in a temperature range from 0°C to +50°C, preferably at +10°C to +30°C. The reaction may take place under atmospheric, under elevated or under reduced pressure (e.g. from 0.5 to 5 bar); it is usual to operate under atmospheric pressure.
The compounds of the formulae (II) and (III) are sub-quantities of the compounds of the formulae (IIa) and (IIIa) respectively, where R35 is methyl. The preparation of the compounds (IIa) and (IIIa) takes place in analogy to the preparation of the compound of the formulae (II) and (III) as described above.
The above-described processes are illustrated by way of example by the following synthesis schemes (Scheme 3 to 13, 18): Scheme 3 H C O H C CH 3 3 3 CH 3 N O H H N N N O HN N O O CH O CH O O 3 CH 3 3 x TFA H C CH CH 3 3 1. N H a) O 2. Pd/C H C CH H C 3 3 CH O 3 N H N N N O H N N 2 N O O CH O CH O O CH 3 3 3 H C CH CH 3 3 O O N O b) CH H C CH H C 3 3 CH 3 N O O H H N N N O N N N O O CH O CH O O 3 3 CH H C CH CH 3 O 3 3 3 [a): 1. Water/dioxane, 1N HCl, 100°C; 2. H2, Pd/C, methanol, RT; b): NaHCO3, H2O, dioxane, RT]. 2012/075277 Scheme 4 H C CH H C 3 3 CH 3 NH2 O H H N H N N N 2 O N N O O CH3 O CH O O 3 CH3 H C CH CH3 3 3 N O a) N O + O O N H3C CH3 H C 3 CH3 NH O H 2 H H N O N N N O N N O O HN CH O CH3 O O CH 3 3 O H C CH CH 3 3 [a): Diisopropylethylamine, DMF, RT]. 2012/075277 Scheme 5 H C CH H C 3 3 CH O 3 N H N N N O HN N O O CH O CH O O CH 3 3 3 x TFA H C CH CH 3 3 a) HO NaCNBH3 H C CH H C 3 CH 3 3 O 3 N H N HO N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 O NH b) N O CH N H C CH H C O 3 CH O 3 N N 3 H N O N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 [a): Water/dioxane, 1N HCl, 100°C; b): HATU, diisopropylamine, DMF, RT].
Scheme 6 H C CH H3C 3 3 CH 3 N O H H N HO N N O N N O O O CH O CH O O 3 CH 3 3 H3C CH3 CH 1. HN O OH a) O CH 2. Pd/C Boc H C CH H C O HN 3 3 CH N O 3 H H N HO O N N O N N O O O CH O O CH O O CH 3 CH 3 3 CH3 3 H C CH 3 3 1. N NH x HCl b) O 2. TFA H C CH H3C O 3 CH NH2 3 O 3 N H H N N O N N O HN N N O O O CH O CH O CH O O 3 CH O 3 3 3 H C CH CH 3 3 [a): 1. EDCI, DMAP, dichloromethane, RT; 2. H2, methanol, RT ; b): 1. EDCI, HOBt, ropylamine, DMF, RT; 2. dichloromethane, RT].
Scheme 7 H C CH H3C CH 3 3 3 N O H H N HO N N O N N O O O CH O CH O O 3 3 CH CH 3 H C CH 3 3 3 1. Z N OH 2. Pd/C H C CH H C 3 3 CH N O 3 H H N O N N O N N O O HN O CH O CH3 O O CH 3 3 H C CH CH3 3 3 b) O H C CH H C 3 3 CH O 3 N H N O N N O N N O O O N O CH O CH3 O O CH 3 3 H C CH CH3 3 3 [a): 1. EDCI, DMAP, dichloromethane, RT; 2. H2, ol, RT ; b): HATU, diisopropylamine, DMF, RT].
WO 87716 Scheme 8 H C CH H C 3 3 CH O 3 O H N HO N N O N N O O O CH O CH O O 3 3 CH CH 3 H C H C CH 3 3 3 3 H C CH H C 3 3 CH O O O 3 H N O N N O N N N O O O CH O CH O O CH 3 3 3 H C O H C CH CH 3 3 3 3 [a): EDCI, dichloromethane, RT]. 2012/075277 Scheme 9 H C CH H C 3 CH 3 3 3 O O H H N HO N N NH2 N N O O O CH O CH3 O O CH 3 3 H C CH CH3 3 3 a) Bn 2. Pd/C H C CH H C 3 CH 3 3 3 O O H H H N N N N NH2 N N O O O O CH O CH3 O O CH 3 3 H C CH CH3 3 3 OH N H C CH H3C 3 CH 3 O 3 O H H N N N N NH2 N N O O O O CH O CH O O 3 CH 3 CH 3 O H3C CH3 3 O N N H [a): HATU, diisopropylethylamine, DMF, RT; 2. H2, methanol, RT; b): EDCI, DMAP, dichloromethane, RT].
WO 87716 Scheme 10 H C CH H3C 3 CH 3 3 O O H H N HO N N N N N H O O O CH O CH3 O O CH 3 3 H C CH CH3 3 3 CH O H3C 3 1. NH2 H3C O a) CH 2. TFA H3C CH3 H C 3 CH3 O O O H H H N N N N N HO N N H O O H C O CH O CH O O CH 3 3 3 3 H3C CH3 CH O H C CH H C 3 CH 3 3 O O O H H H N N N N N N O N N H O O O H C O CH3 O CH O O 3 CH3 H C CH CH 3 3 [a): 1. HATU, diisopropylethylamine, DMF, RT; 2. dichloromethane, RT; b): EDCI, DMAP, dichloromethane, RT]. 2012/075277 Scheme 11 H C CH H C 3 3 CH 3 N O H H N N N O H N N 2 N O O CH O CH O O 3 3 CH H CH CH 3 C 3 3 3 O O O O a) N N O O O O H C CH H C 3 CH N O O 3 3 O 3 H N N N O O N N N H O O N CH O CH O O 3 CH O 3 O H C CH CH 3 3 3 [a): Diisopropylethylamine, DMF, RT].
Scheme 12 H C CH H C 3 3 CH N O 3 H H N HO N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 H C O O 3 OH 1. H C 3 CH HN 3 CH3 2. TFA H C CH H C 3 CH 3 3 N O 3 H N O N N O H N N N 2 O O CH O CH O CH O O CH 3 3 3 H C CH CH 3 3 3 x TFA O O O O b) N N O O O O H C CH H C O 3 3 CH O 3 N H N O N N O HN N N O O CH O CH O CH O O CH O 3 3 3 3 H C CH CH 3 3 O O [a): 1. EDCI, DMAP, dichloromethane, RT; 2. romethane, RT; b): diisopropylamine, DMAP, dichloromethane, RT]. 2012/075277 Scheme 13 H C CH H3C 3 CH 3 O 3 N H N O N N O N N O O HN O CH O CH O O CH 3 3 H3C CH3 CH 3 O O O O a) N N O O O O H C CH H C 3 CH 3 3 N O H H N O N N O N N O O O N O CH O CH3 O O CH 3 3 H C CH CH3 3 3 [a): DMAP, diisopropylamine, dichloromethane, RT].
Scheme 18 H C CH H C O 3 CH 3 3 O 3 N H N N N O HN N x TFA O O CH O CH O O 3 3 H C H C CH 3 3 H C CH 3 3 3 1. H HN NaCNBH3 a) Z O 2. Pd/C C CH C O H H 3 3 CH N O 3 H N H N N N 2 O N N O O CH O CH O O 3 3 H C H C CH 3 3 H C CH 3 3 3 H C CH H C 3 3 CH 3 N O H H N N N O N N O O O NH CH O CH O O 3 3 H C H C 3 3 O H C CH CH 3 3 [a): 1. dioxane, 1N HCl, 100°C; 2. H2, Pd/C, methanol, RT; b): HATU, diisopropylethylamine, RT].
The compounds of the formula (IV) can be prepared from commercially available amino acid building blocks or those known from the literature (see, for e, Pettit et al., Synthesis 1996, 719; Shioiri et al., Tetrahedron Lett. 1991, 32, 931; Shioiri et al., Tetrahedron 1993, 49, 1913; Koga et al., Tetrahedron Lett. 1991, 32, 2395; Vidal et al., Tetrahedron 2004, 60, 9715; Poncet et al., Tetrahedron 1994, 50, 5345. Pettit et al., J. Org. Chem. 1994, 59, 1796) in analogy to processes known from the literature, in accordance with customary methods of peptide chemistry, and as described in the present mental section. The synthesis schemes below (Scheme 14 to 16) illustrate the preparation by way of example.
Scheme 14 OMe OH H O O N H N O O N O H N H C O 2 O 3 H C O a) 3 b) 3 CH H C 3 3 CH [ a): Hydroxylamine hydrochloride, KOH, MeOH, 0°C → RT; b): BrCH2(CH2)2CH2Br, K2CO3, acetone, ].
Scheme 15 3 1. H CH O N 3 H C OH 3 H C O O H C CH O CH 3 3 H N O CH HN 3 2 3 CH N 3 CH CH O O CH 2. H2, Pd/C 3 3 CH O O CH CH 3 3 3 H C CH CH 3 3 3 [a): 1. Diisopropylethylamine, BEP, dichloromethane, -10°C → RT; 2. MeOH].
Scheme 16 H C CH CH OH H C 3 H N O CH N 2 3 N 3 CH H C 3 3 O O O CH O O CH O CH H C CH 3 CH 3 3 3 3 O O 1. 1. N H C CH 3 3 FmocH C OH b) 3 a) H N 2 x 2 TFA CH3 O 2. TFA 2. TFA H C CH H C 3 3 3 CH 3 N O H H N FmocH C N OH + 3 N O N N H O O CH O CH O O x TFA 3 CH H C CH 3 CH 3 3 3 3 H C CH H C CH 3 3 3 3 N O H H N N N O HN N O O CH O CH O O 3 H C CH 3 CH CH 3 3 3 3 [ a): 1. Diisopropylethylamine, BEP, DMF, RT; 2. dichloromethane; b): 1. HATU, ropylethylamine, DMF, RT; 2. romethane, RT; c): 1. HATU, diisopropylethylamine, DMF, RT; 2. piperidine, DMF, RT].
The compounds of the formulae (XI), (XIII), (XV), (XVII) and (XXI), including, where appropriate, chiral or diastereomeric forms thereof, are available commercially or are bed as such in the literature, or they can be prepared by routes that are obvious to the skilled person, in analogy to methods published in the literature. Numerous comprehensive instructions and also literature information on the preparation of the starting materials are also given in the experimental n, in the section relating to the preparation of the starting compounds and intermediates.
The compounds of the formulae (V), (VII), (VIII), (X), (XVIII), (XX) and (XXIII), including, where appropriate, chiral or diastereomeric forms f, are known from the literature, or can be prepared by routes which are obvious to the skilled person, in analogy to methods published in the literature. Numerous comprehensive instructions and also literature ation on the preparation of the starting materials are also given in the experimental section, in the section relating to the preparation of the starting compounds and intermediates.
Alternatively, individual steps of the preparation ce may be d out in a different order or with other protective group combinations. This approach is illustrated by way of example in the synthesis schemes below (Scheme 17, 19, 20 and 21). 2012/075277 Scheme 17 H CH O H N H C OH HN 3 N HN OH O H C CH 3 3 O O O O H C CH 3 3 CH CH 3 3 O 3 H C CH H C 3 3 O 3 H N O CH 2 3 H N CH N OH CH O O CH N N H C CH 3 CH 3 3 3 HN O CH O CH O O 3 H C CH 3 CH 3 3 3 3 N N O c) O O x TFA H C CH H C 3 3 O 3 CH 3 N H N N N O N N O O HN O CH O CH O O 3 CH H C CH 3 CH 3 3 3 3 1. H2 O O N O O H C CH H C 3 3 CH N O 3 3 H N N N N O N N O O O CH O CH O O 3 CH H C CH 3 CH 3 3 3 3 [ a): Borane-pyridine complex, acetic acid, MeOH; b): 1. HOBt, EDCI, diisopropylethylamine, DMF, RT; 2. TFA, dichloromethane, RT; c): HATU, diisopropylethylamine, DMF, RT; d): 1.
Pd/C, MeOH, RT; 2. NaHCO3, dioxane, water].
Scheme 19 H3C CH3 CH 1. CH 3 OH 3 O N H C H C 3 CH H C O CH3 O O 3 O 3 3 CH H N O CH CH 3 H 2 3 N OH N CH O N N CH O O CH 3 3 CH O CH O O H C CH CH a) 3 3 3 H C 3 CH 3 H C CH CH 3 3 CH 3 3 3 N b) O O 3 CH H C 3 3 CH O O 3 H O N N O N N O O CH O CH O O CH H C 3 CH 3 CH 3 3 CH CH 3 H C 3 3 3 2. O H C CH H C 3 O 3 CH NH 3 3 2 H N N N O HN N O O CH O CH O O 3 H C CH 3 CH CH 3 3 3 3 [ a): 1. HATU, diisopropylethylamine, DMF, RT; 2. TFA, dichloromethane, RT; 3.
((H3C)3C(C=O))2O, DMF, diisopropylethylamine; b): diisopropylethylamine, BEP, DMF, RT; c): 1. H2, Pd/C (10%), methanol, RT; 2. HATU, diisopropylethylamine, DMF, RT; 3. TFA, dichloromethane, RT].
Scheme 20 N CH O H H C OH HN N H C CH3 HN OH O 3 O O O O H C CH a) 3 3 CH3 CH 1. 3 O 3 H C CH H C 3 3 O 3 H2N O CH 3 H N CH3 N OH CH3 O O CH N N H3C CH3 CH 3 3 NH CH O CH O O 2 3 H C CH 3 CH 3 3 3 O O 3 N O O O 2. H 3 N OH O N N CH O O c) O H C CH CH 3 3 3 CH3 N N O d) O O x CF3 COOH H H C O 3 CH 3 N O O H H N N N O N N O O CH O O CH 3 O H C CH CH 3 3 3 3 [a): Borane-pyridine complex, acetic acid, MeOH; b): 1. HOBt, EDCI, diisopropylethylamine, DMF, RT; 2. TFA, dichloromethane, RT; 3. H2, Pd/C, MeOH, RT c): 1. , dioxane, water; d): HATU, diisopropylethylamine, DMF, RT]. 2012/075277 Scheme 21 H N O CH 2 N 3 CH O O CH H C CH 3 CH 3 3 3 z O 1. N H C OH 3 a) H C CH 3 3 2. H b) CH 2 3 CH CH OH C 3 3 H N H C CH H C 3 3 H C O 3 3 O O O O CH H CH 3 O 3 HN N CH CH O CH O O CH 3 NH H C CH 3 CH 3 O O 3 3 3 1. N e) 2 x TFA H c) 2. TFA O d) H C CH H C 3 3 O 3 CH 3 N H N BnO N OH N O N N H + O O O CH O CH O O 3 CH H C CH 3 CH x TFA 3 3 3 3 H C CH H C 3 3 CH O 3 3 N H N HO N N O N N O O O CH O CH O O 3 CH H C CH 3 CH 3 3 3 3 [a): 1. HOBt, EDCI, diisopropylethylamine, DMF, RT; b) H2, Pd/C, MeOH, RT; c) boranepyridine complex, acetic acid, MeOH; d) TFA, dichloromethane, RT; e): HATU, diisopropylethylamine, DMF, RT; f): HATU, diisopropylethylamine, DMF, RT; g) H2, Pd/C, MeOH, RT].
WO 87716 The FGFR2 cancer target molecule of the binder of the present ion is known to the skilled person. The full-length FGFR2 is identified as FGFR2 alpha (SEQ ID NO: 1), while the isoform lacking the D1 domain is identified as FGFR2 beta (SEQ ID NO: 2) (see Figure 1). Alternative splicing in domain 3 leads to two different variants, namely FGFR2 IIIb, which is encoded by the exons 7 and 8, and FGFR2 IIIc, which is encoded by the exons 7 and 9 (see Figure 1).
In one embodiment of the ion the binder binds – preferably specifically – to FGFR2. In a further subject of the invention, the binder binds – preferably specifically – to the extracellular domain of the target molecule FGFR2 (see Figure 1).
In a further embodiment of the invention, the binder binds – preferably specifically – to one or more forms of the human FGFR2 polypeptide. In a further subject of the invention, the binder binds – preferably specifically – to all isoforms and splice variants of FGFR2. In the text below, the concept of different “forms” of FGFR2 includes, though is not limited to, different isoforms, different splice variants, different glycoforms or FGFR2 polypeptides which o different translational and post-translational modifications.
In a r embodiment of the invention, the binder binds - preferably specifically – to the N- terminal domains of the cancer target molecule FGFR2. In a further t of the invention, the binder binds – preferably specifically – to the extracellular N-terminal epitope SLVEDTTLEPE 15 ) of FGFR2 (SEQ ID NO: 23).
In a further embodiment of the invention, the binder also binds – preferably specifically – to the FGFR2 of different s. The effect of this is that the conjugates of the invention can more easily, pharmacologically, be investigated in these s. Preferred species are rodents, more particularly mice or rats, but also dogs, pigs and non-human es.
In one preferred embodiment the binder, after binding to FGFR2 on the target cell, is internalized by the target cell as a result of the binding. The effect of this is that the binder-drug conjugate, which may be an immunoconjugate or an ADC, is taken up by the target cell.
In one embodiment the binder is a binding n. In one red embodiment the binder is an antibody, an antigen-binding antibody fragment, a pecific antibody or an antibody mimetic.
Preferred antibody mimetics are affibodies, adnectins, anticalins, DARPins, avimers, or nanobodies. Preferred multispecific antibodies are bispecific and cific antibodies.
In one preferred embodiment the binder is an antibody or an antigen-binding antibody fragment, more preferably an isolated antibody or an isolated antigen-binding antibody fragment.
Preferred antigen-binding antibody fragments are Fab, Fab’, F(ab’)2 and Fv fragments, diabodies, DAbs, linear antibodies and scFv. Particularly preferred are Fab, ies and scFv.
In one particularly preferred embodiment the binder is an antibody. Particularly preferred are monoclonal antibodies or antigen-binding antibody fragments thereof. Further particularly preferred are human, humanized or chimeric antibodies or antigen-binding antibody fragments thereof.
In one preferred embodiment, the antibody or the antigen-binding fragment comprises the amino acid sequence of the CDR sequences of the variable light and heavy chain of the antibody M048- D01-hIgG1.
In a further red embodiment, the dy or the antigen-binding fragment comprises the amino acid sequence of the CDR sequences of the variable light and heavy chain of the antibody M048-D01-hIgG1 represented in SEQ ID NO: 15 (H-CDR1), SEQ ID NO: 16 (H-CDR2), SEQ ID NO: 17 (H-CDR3), SEQ ID NO: 18 (L-CDR1), SEQ ID NO: 19 2) and SEQ ID NO: 20 (L-CDR3).
In a further preferred embodiment, the dy or the antigen-binding fragment comprises the amino acid sequence of the variable light and heavy chains of the dy M048-D01-hIgG1 or M048-D01-hIgG1-b.
In a further preferred embodiment, the antibody or the n-binding fragment comprises the amino acid sequence of the variable light and heavy chains of the antibody M048-D01-hIgG1, represented in SEQ ID NO: 12 (Vl) and SEQ ID NO: 11 (Vh), or of the variable light and heavy chains of the antibody 01-hIgG1-b, ented in SEQ ID NO: 14 (Vl) and SEQ ID NO: 13 (Vh).
In a further particularly preferred embodiment, the antibody or the antigen-binding fragment comprises the amino acid sequence of the variable light and heavy chain of the antibody M048- D01- hIgG1-b represented in SEQ ID NO: 14 (Vl) und SEQ ID NO: 13 (Vh).
In a further particularly red embodiment, the antibody comprises the amino acid ce of the light and heavy chain of the antibody M048-D01-hIgG1-b represented in SEQ ID NO: 9 (light chain) and SEQ ID NO: 10 (heavy chain).
In a r particularly preferred embodiment, the antibody comprises the amino acid sequence of the light and heavy chain of the antibody M048-D01-hIgG1 represented in SEQ ID NO: 7 (light chain) and SEQ ID NO: 8 (heavy chain).
Examples of further FGFR2 antibodies are the GAL-FR21-mIgG1 (SEQ ID NO: 3 and SEQ ID NO: 4) and GAL-FR22-mIgG2a (SEQ ID NO: 5 and SEQ ID NO: 6) antibodies that are described in this invention. The two last-mentioned antibodies were constructed on the basis of WO2010/054265 from the variable regions, described therein, of the light (Vl) and heavy (Vh) chains of the antibodies GAL-FR21 (SEQ ID NO: 1 and SEQ ID NO: 4 from WO2010/054265) and GAL-FR22 (SEQ ID NO: 7 and SEQ ID NO: 8 from WO2010/054265), with the variable regions of GAL-FR21 having been reformatted into an mIgG1 format, while the variable regions of 22 were reformatted into an mIgG2a format.
Antibodies or antigen-binding antibody fragments which bind cancer target molecules may be prepared by a person of ordinary skill in the art using known processes, such as, for e, chemical synthesis or recombinant expression. Binders for cancer target molecules may be ed commercially or may be prepared by a person of ordinary skill in the art using known processes, such as, for example, al synthesis or recombinant expression. Further processes for preparing dies or antigen-binding antibody fragments are described in WO2007070538 (see page 22 “Antibodies”). The skilled person knows how processes such as phage display libraries (e.g. Morphosys HuCAL Gold) can be compiled and used for discovering antibodies or antigen-binding antibody fragments (see WO2007070538, page 24 ff and Example 1 on page 70, Example 2 on page 72). Further processes for ing antibodies that use DNA libraries from B-cells are described for example on page 26 (WO2007070538). Processes for humanizing antibodies are described on page 30-32 of WO2007070538 and in detail in Queen, et al., Pros.
Natl. Acad. Sci. USA 86:10029-10033, 1989 or in WO90/0786. Furthermore, processes for the recombinant expression of proteins in general and of antibodies in particular are known to the skilled person (see, for example, in Berger and Kimrnel (Guide to Molecular Cloning Techniques, s in Enzymology, Vo1. 152, Academic Press, Inc.); Sambrook, et al., (Molecular g: A Laboratory , (Second Edition, Cold Spring Harbor Laboratory Press; Cold Spring Harbor, N.Y.; 1989) Vol. 1-3); Current Protocols in Molecular Biolony, (F.M. l et al.
, Current Protocols, Green Publishing Associates, Inc./John Wiley & Sons, Inc.); Harlow et al., (MonocIonal Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press (19881, Paul [Ed.]); Fundamental Immunology, (Lippincott ms & Wilkins (1998)); and , et al., (Using Antibodies: A tory Manual, Cold Spring Harbor Laboratory Press (1998)). The skilled person knows the corresponding vectors, promoters and signal peptides which are necessary for the expression of a protein/antibody. Commonplace processes are also described in WO2007070538 on pages 41–45. Processes for preparing an IgG1 antibody are described for e in WO2007070538 in Example 6 on page 74 ff. Processes which allow the determination of the alization of an antibody after binding to its antigen are known to the skilled person and are bed for example in WO2007070538 on page 80. The skilled person is able to use the processes described in WO2007070538 that have been used for preparing carboanhydrase IX (Mn) antibodies in y for the preparation of antibodies with different target molecule specificity.
Further examples of FGFR2 binders are the single chain Fv antibody fragments PRO-007 (binds FGFR2 with high affinity) and PRO-001 (binds FGFR3 with high affinity and FGFR2 with low affinity) described in WO2007144893.
The compounds of the invention s le pharmacological properties and can be used for the prevention and treatment of diseases in humans and animals.
The binder-drug conjugates (ADCs) of the invention, of the formula (Ia), exhibit a high and specific cytotoxic activity with regard to tumour cells, as may be shown on the basis of the assays set out in the present experimental section (Section C). This high and specific cytotoxic activity on the part of the binder-drug conjugates (ADCs) of the invention, of the formula (Ia), is achieved h the appropriate combination of the new N,N-dialkylauristatin derivative and binder with linkers which exhibit not only an enzymatically, hydrolytically or reductively cleavable predetermined break point, for the release of the toxophores, but also no such predetermined break point. More ularly, through the use of stable linkers which have no enzymatically, hydrolytically or ively cleavable predetermined break point for the release of the toxophores, and which, following uptake of the ADCs into the tumour cell and following complete intracellular, enzymatic breakdown of the antibody, still remain wholly or partly intact, the activity is confined very ically to the tumour cell. Compatibility between ADCs and stable linkers poses, among other things, that the metabolites formed ellularly can be formed with sufficient efficacy, are able to reach their target and are able there to develop their antiproliferative activity on the target with sufficient potency, without being carried out of the tumour cell again hand by transporter proteins. Such a compatibility of the ADCs with a stabile linker chemistry and with the target in question offers an enlarged therapeutic window (see, e.g., L. Ducry, Bionconjugate Chem. 2010, 21-5; A.G. Polson, Cancer Res. 2009, 69, 2358).
More particularly, the binder-drug conjugates of the invention, of the formula (Ia), exhibit a high and specific cytotoxic activity with respect to tumour cells which express FGFR2. The activity with respect to tumour cells which do not express FGFR2 is significantly weaker at the same time.
On the basis of this profile of ties, the nds of the invention are therefore suitable to a particular degree for the treatment of hyperproliferative diseases in humans and in mammals generally. The nds are able on the one hand to inhibit, block, reduce or lower cell proliferation and cell division, and on the other hand to se apoptosis.
The hyperproliferative diseases for the treatment of which the compounds of the invention can be employed include in particular the group of cancer and tumour es. In the context of the present invention, these are understood as meaning, in ular, the following diseases, but without being limited to them: y carcinomas and mammary tumours (ductal and lobular forms, also in situ, triple-negative, egative), tumours of the atory tract (parvicellular and non-parvicellular oma, bronchial oma), cerebral tumours (e.g. of the brain stem and of the alamus, astrocytoma, oblastoma, ependymoma and neuro-ectodermal and pineal tumours), tumours of the digestive organs (oesophagus, carcinomas of the oesophagogastric junction (=EGJ), stomach (diffuse and intestinal forms), gall bladder, small intestine, large intestine, rectum), liver tumours (including hepatocellular carcinoma, cholangiocellular carcinoma and mixed hepatocellular and cholangiocellular carcinoma), tumours of the head and neck region (larynx, hypopharynx, nasopharynx, oropharynx, lips and oral cavity), skin s (squamous epithelial carcinoma, Kaposi sarcoma, malignant melanoma, Merkel cell skin cancer and nonmelanomatous skin cancer), tumours of soft tissue (including soft tissue sarcomas, osteosarcomas, malignant fibrous histiocytomas, lymphosarcomas and rhabdomyosarcomas), tumours of the eyes (including intraocular melanoma and retinoblastoma), tumours of the endocrine and exocrine glands (e.g. thyroid and parathyroid glands, pancreas and salivary gland), tumours of the urinary tract (tumours of the bladder, penis, kidney, renal pelvis and ureter) and tumours of the uctive organs (carcinomas of the endometrium, cervix, ovary, vagina, vulva and uterus in women and omas of the prostate and testicles in men). These also include proliferative blood diseases in solid form and as circulating blood cells, such as lymphomas, leukaemias and myeloproliferative diseases, e.g. acute myeloid, acute lymphoblastic, chronic lymphocytic, chronic myelogenic and hair cell leukaemia, and also orrelated lymphomas, Hodgkin's lymphomas, dgkin's lymphomas, cutaneous T-cell lymphomas, Burkitt's lymphomas and lymphomas in the central nervous system.
Preferred hyperproliferative diseases for anti-FGFR2 binder-drug conjugates Hyperproliferative diseases for the ent of which the compounds of the invention can be preferably employed are FGFR2-expressing tumours, such as, for example, stomach oma (intestinal and diffuse types), signet ring oma, especially of diffuse type, oesophageal cancer, cancer of the oesophagogastric junction (EGJ), breast cancer, cancer of the large intenstine, colorectal carcinoma, rectal carcinoma, te cancer, kidney cancer, carcinomas of the head and neck region, pancreatic cancer, liver cancer, cervical carcinoma, ovarian carcinoma, endometrial carcinoma, more particularly of trioid type, of papillary serous type, or of clear cell subtype, lung cancer, more particularly non-small-cell lung carcinoma (NSCLC), adenocarcinoma, us carcinoma and pancreatic carcinoma.
These well-described diseases in humans can also occur with a comparable aetiology in other mammals and can be treated there with the compounds of the present invention.
In the context of this ion, the term "treatment" or "treat" is used conventionally and means the care, management and support of a patient with the aim of combatting, shing, attenuating or relieving a disease or health defect and of ing the living ions which are adversely affected by this e, such as in the case of a cancer disease.
The present ion thus further provides the use of the compounds of the invention for the treatment and/or prevention of diseases, in particular the abovementioned es.
The present invention furthermore provides the use of the compounds of the invention for the preparation of a medicament for the treatment and/or prevention of diseases, in particular the abovementioned diseases.
The present invention furthermore provides the use of the compounds of the invention in a method for the treatment and/or prevention of diseases, in particular the abovementioned es.
The present invention furthermore provides a method for the treatment and/or prevention of diseases, in particular the abovementioned diseases, using an effective amount of at least one of the compounds of the invention.
The compounds according to the invention can be employed by themselves or, if required, in combination with one or more other pharmacologically active substances, as long as this combination does not lead to undesirable and ptable side effects. The present invention furthermore therefore es medicaments comprising at least one of the compounds of the invention and one or more further drugs, in particular for the treatment and/or prevention of the abovementioned diseases.
For e, the compounds of the present invention can be combined with known antihyperproliferative, cytostatic or cytotoxic nces for the treatment of cancer diseases.
Suitable drugs in the combination which may be mentioned by way of example are as follows: eukin, alendronic acid, alfaferone, alitretinoin, allopurinol, aloprim, aloxi, altretamine, aminoglutethimide, amifostine, amrubicin, ine, anastrozole, anzmet, aranesp, arglabin, arsenic trioxide, aromasin, 5-azacytidine, azathioprine, BCG or CG, bestatin, betamethasone acetate, betamethasone sodium phosphate, bexarotene, bleomycin sulphate, broxuridine, bortezomib, busulfan, calcitonin, campath, capecitabine, carboplatin, casodex, ne, celmoleukin, cerubidin, chlorambucil, cisplatin, cladribin, clodronic acid, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunoxome, decadron, on phosphate, delestrogen, denileukin diftitox, depomedrol, deslorelin, dexrazoxane, diethylstilbestrol, diflucan, docetaxel, doxifluridine, doxorubicin, dronabinol, DW-166HC, eligard, elitek, ellence, emend, epirubicin, n-alfa, epogen, eptaplatin, ergamisol, e, estradiol, estramustine sodium phosphate, ethinylestradiol, ethyol, etidronic acid, etopophos, etoposide, fadrozole, farstone, filgrastim, finasteride, fligrastim, floxuridine, fluconazole, fludarabin, 5-fluorodeoxyuridine monophosphate, -fluorouracil (5-FU), fluoxymesterone, ide, formestane, fosteabine, fotemustine, fulvestrant, gammagard, gemcitabine, gemtuzumab, gleevec, gliadel, goserelin, granisetron hydrochloride, histrelin, hycamtin, hydrocortone, erythro-hydroxynonyladenine, hydroxyurea, ibritumomab tiuxetan, idarubicin, ifosfamide, eron-alpha, interferon-alpha-2, eron- alpha-2α, interferon-alpha-2β, interferon-alpha-n1, interferon-alpha-n3, interferon-beta, interferongamma-1α , interleukin-2, intron A, iressa, ecan, kytril, lentinan sulphate, letrozole, leucovorin, leuprolide, leuprolide acetate, levamisole, levofolic acid calcium salt, levothroid, levoxyl, lomustine, lonidamine, marinol, mechlorethamine, mecobalamin, medroxyprogesterone acetate, megestrol acetate, melphalan, menest, 6-mercaptopurine, mesna, methotrexate, metvix, osine, minocycline, mitomycin C, mitotane, mitoxantrone, modrenal, myocet, atin, neulasta, a, neupogen, mide, nolvadex, NSC-631570, OCT-43, octreotide, ondansetron hydrochloride, orapred, oxaliplatin, paclitaxel, pediapred, pegaspargase, s, pentostatin, picibanil, pilocarpine hydrochloride, pirarubicin, plicamycin, porfimer sodium, prednimustine, prednisolone, prednisone, in, procarbazine, t, raltitrexed, rebif, rhenium-186 etidronate, rituximab, roferon-A, romurtide, salagen, sandostatin, sargramostim, semustine, sizofiran, sobuzoxane, solu-medrol, streptozocin, strontium-89 de, synthroid, tamoxifen, tamsulosin, tasonermin, tastolactone, taxoter, teceleukin, temozolomide, side, testosterone propionate, testred, thioguanine, thiotepa, thyrotropin, onic acid, topotecan, toremifen, tositumomab, tastuzumab, teosulfan, tretinoin, l, trimethylmelamine, trimetrexate, triptorelin acetate, triptorelin pamoate, UFT, uridine, valrubicin, vesnarinone, vinblastine, vincristine, vindesine, vinorelbine, virulizin, zinecard, zinostatin-stimalamer, zofran; ABI-007, acolbifen, actimmune, affinitak, aminopterin, arzoxifen, asoprisnil, tane, atrasentan, n, BAY 43-9006 (sorafenib), CCI-779, 1, celebrex, cetuximab, crisnatol, cyproterone acetate, decitabine, DN-101, doxorubicin-MTC, dSLIM, dutasteride, edotecarin, eflornithine, an, fenretinide, histamine dihydrochloride, histrelin el implant, holmium-166 DOTMP, onic acid, interferon-gamma, intron-PEG, ixabepilone, keyhole limpet hemocyanine, L- 651582, lanreotide, lasofoxifen, libra, lonafarnib, miproxifen, minodronate, MS-209, liposomal MTP-PE, MX-6, nafarelin, bicin, neovastat, nolatrexed, oblimersen, onko-TCS, osidem, paclitaxel polyglutamate, pamidronate disodium, PN-401, QS-21, quazepam, R-1549, raloxifen, ranpirnas, 13-cis -retic acid, satraplatin, seocalcitol, T-138067, tarceva, exin, thymosinalpha-1 , tiazofurin, tipifarnib, tirapazamine, TLK-286, toremifen, transMID-107R, valspodar, vapreotide, nib, verteporfin, vinflunin, Z-100, onic acid and combinations of these.
In a preferred embodiment, the compounds of the present invention can be combined with antihyperproliferative agents, which can be, by way of example – without this list being conclusive as follows: aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine, bleomycin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, lstilbestrol, 2',2'-difluorodeoxycytidine, docetaxel, doxorubicin (adriamycin), epirubicin, epothilone and its derivatives, erythro-hydroxynonyladenin, ethinylestradiol , etoposide, fludarabin phosphate, 5-fluorodeoxyuridine, 5-fluorodeoxyuridine monophosphate , rouracil, fluoxymesterone, flutamide, hexamethylmelamine, hydroxyurea, hydroxyprogesterone caproate, idarubicin, ifosfamide, interferon, irinotecan, orin, lomustine, rethamine, medroxyprogesterone acetate, megestrol acetate, melphalan, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitotane, mitoxantrone, paclitaxel, pentostatin, N-phosphonoacetyl L-aspartate (PALA), ycin, prednisolone, prednisone, bazine, raloxifen, semustine, ozocin, tamoxifen, teniposide, testosterone propionate, thioguanine, thiotepa, can, trimethylmelamine, uridine, stine, vincristine, vindesine and vinorelbine.
The compounds of the invention can also be combined in a very promising manner with biological therapeutics such as antibodies (e.g. avastin, rituxan, erbitux, herceptin). The nds of the invention can also achieve positive effects in combination with therapies directed against angiogenesis, such as, for example, with avastin, ib, recentin, regorafenib, sorafenib or nib. Combinations with inhibitors of the proteasome and of mTOR and also with antihormones and steroidal metabolic enzyme inhibitors are likewise particularly suitable because of their favourable profile of side effects.
Generally, the following aims can be pursued with the combination of compounds of the present invention with other agents having a cytostatic or cytotoxic action: • an ed activity in slowing down the growth of a tumour, in reducing its size or even in its complete elimination compared with treatment with an individual drug; • the possibility of employing the chemotherapeutics used in a lower dosage than in monotherapy; • the possibility of a more tolerable therapy with few side effects compared with individual administration; • the possibility of ent of a broader spectrum of tumour diseases; 2012/075277 • the achievement of a higher rate of response to the therapy; • a longer survival time of the patient compared with present-day standard therapy.
The compounds according to the invention can moreover also be employed in combination with radiotherapy and/or surgical intervention.
The present invention furthermore provides medicaments which comprise at least one compound of the invention, conventionally together with one or more inert, xic, ceutically suitable ents, and the use thereof for the abovementioned purposes.
The compounds of the invention can act ically and/or locally. They can be administered in a suitable manner for this purpose, such as for example parenterally, possibly by means of inhalation, or as an implant or stent.
The compounds of the invention can be stered in suitable administration forms for these administration routes.
Parenteral administration can be effected with bypassing of an absorption step (e.g. intravenously, intraarterially, intracardially, intraspinally or intralumbally) or with inclusion of an tion (e.g. intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally).
Administration forms which are suitable for parenteral administration include injection and infusion formulations in the form of solutions, suspensions, emulsions or lizates. Parenteral administration is preferred, in particular intravenous stration.
In general, it has proved advantageous in the case of parenteral administration to administer amounts of from about 0.001 to 1 mg/kg, preferably about 0.01 to 0.5 mg/kg of body weight to achieve effective results. heless it may be necessary to deviate from the amounts mentioned, and in particular depending on the body weight, administration route, individual behaviour towards the active nd, nature of the formulation and point of time or interval at which administration takes place. Thus in some cases it may be sufficient to manage with less than the abovementioned minimum amount, while in other cases the upper limit mentioned must be exceeded. In the case where relatively large amounts are administered, it may be advisable to distribute these into several individual doses over the day.
The following es illustrate the invention. The invention is not d to the examples.
The percentage figures in the following tests and examples are percentages by weight, unless stated otherwise; parts are parts by weight. Solvent , dilution ratios and concentration data of liquid/liquid solutions in each case relate to the volume.
A. Examples Abbreviations and acronyms: ABCB1 ATP-binding te sub-family B member 1 (synonym for P-gp and MDR1) abs. te ADC antibody-drug-conjugate Ac acetyl aq. aqueous, aqueous solution ATP adenosine triphosphate BCRP breast cancer resistance n, an efflux transporter Boc tert -butoxycarbonyl br. broad (in NMR) Ex. example ca . circa , approximately CAIX carboanhydrase IX CI chemical ionization (in MS) d doublet (in NMR) d day(s) TLC thin-layer chromatography DCI direct chemical ionization (in MS) dd t of a doublet (in NMR) DMAP 4-N,N -dimethylaminopyridine DME methoxyethane DMEM Dulbecco’s modified eagle medium (standardized nutrient medium for cell culture) DMF N,N -dimethylformamide DMSO dimethyl sulphoxide DPBS, D-PBS, PBS Dulbecco's phosphate-buffered saline solution PBS = DPBS = D-PBS, pH 7.4, from Sigma, No. D8537 Composition: 0.2 g KCl 0.2 g KH2PO 4 (anhydrous) 8.0 g NaCl 1.15 g Na2HPO 4 (anhydrous) make up to 1 l with H2O dt doublet of a triplet (in NMR) WO 87716 DTT hiothreitol EDC N' -(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride EGFR epidermal growth factor receptor EI electron impact ionization (in MS) ELISA enzyme-linked immunosorbent assay eq. equivalent(s) ESI electrospray ionization (in MS) ESI-MicroTofq ESI- MicroTofq (name of the mass spectrometer, with Tof = time of flight and q = quadrupole) FCS foetal calf serum FGFR2 fibroblast growth factor or 2 Fmoc (9H-fluorenylmethoxy)carbonyl sat. saturated GTP guanosine 5'-triphosphate h hour(s) HATU O-(7-azabenzotriazolyl)-N,N,N',N' -tetramethyluronium hexafluorophosphate HEPES 4-(2-hydroxyethyl)piperazineethanesulphonic acid HOAc acetic acid HOBt 1-hydroxy-1H-benzotriazole e HOSu N-hydroxysuccinimide HPLC high-pressure, high-performance liquid chromatography IC 50 half-maximum inhibitory concentration i.m. intramuscular, administration into the muscle i.v. intravenous, administration into the vein Kato III human tumour cell line conc. concentrated LC-MS liquid chromatography-coupled mass spectrometry LLC-PK1 cells Lewis lung carcinoma pork kidney cell line L-MDR human MDR1 transfected LLC-PK1 cells m multiplet (in NMR) MDA-MB-231 human tumour cell line MDR1 multidrug ence protein 1 MFM-223 human tumour cell line min minute(s) MS mass spectrometry MTT 3-(4,5-dimethylthiazol yl)-2,5-di phenyl trazolium bromide NCI-H716 human tumour cell line NMM N-methylmorpholine NMP N-methylpyrrolidinone NMR nuclear magnetic resonance spectrometry NMRI mouse strain, originating from Naval Medical Research Institute (NMRI) Nude mice experimental animals NSCLC all cell lung cancer (non-parvicellular bronchial carcinoma) PBS phosphate-buffered saline solution Pd/C palladium on activated carbon P-gp P-glycoprotein, a orter protein PNGaseF enzyme for sugar elimination quant. quantitative (for yield) quart quartet (in NMR) quint quintet (in NMR) Rf retention index (for TLC) RT room temperature Rt retention time (for HPLC) s singlet (in NMR) s.c. subcutaneous, administration beneath the skin SCID mice experimental mice with a severe combined immunodeficiency SNU-16 human tumour cell line SUM52-PE human tumour cell line t t (in NMR) tert ry TFA trifluoroacetic acid THF tetrahydrofuran UV ultraviolet spectrometry v/v volume to volume ratio (of a solution) Z benzyloxycarbonyl WO 87716 HPLC and LC-MS methods: Method 1 (LC-MS): Instrument: Waters Acquity SQD UPLC ; column: Waters Acquity UPLC HSS T3 1.8µ 50 mm x 1 mm; eluent A: 1 l water + 0.25 ml 99% strength formic acid, eluent B: 1 l acetonitrile + 0.25 ml 99% strength formic acid; gradient: 0.0 min 90% A → 1.2 min 5% A → 2.0 min 5% A; flow rate: 0.40 ml/min; oven: 50°C; UV ion: 210-400 nm.
Method 2 (LC-MS): Instrument: ass QuattroPremier with Waters UPLC Acquity; column: Thermo il GOLD 1.9µ 50 mm x 1 mm; eluent A: 1 l water + 0.5 ml 50% strength formic acid, eluent B: 1 l acetonitrile + 0.5 ml 50% th formic acid; gradient: 0.0 min 90% A → 0.1 min 90% A → 1.5 min 10% A → 2.2 min 10% A; flow rate: 0.33 ml/min; oven: 50°C; UV detection: 210 nm.
Method 3 (LC-MS): Instrument: Micromass Quattro Micro MS with HPLC Agilent Series 1100; column: Thermo Hypersil GOLD 3µ 20 mm x 4 mm; eluent A: 1 l water + 0.5 ml 50% strength formic acid, eluent B: 1 l acetonitrile + 0.5 ml 50% th formic acid; gradient: 0.0 min 100% A → 3.0 min 10% A → 4.0 min 10% A → 4.01 min 100% A (flow rate 2.5 ml/min) → 5.00 min 100% A; oven: 50°C; flow rate: 2 ml/min; UV detection: 210 nm.
Method 4 (LC-MS): MS instrument: Micromass ZQ; HPLC instrument: HP 1100 Series; UV DAD; column: Phenomenex Gemini 3µ 30 mm x 3.00 mm; eluent A: 1 l water + 0.5 ml 50% strength formic acid, eluent B: 1 l acetonitrile + 0.5 ml 50% strength formic acid; gradient: 0.0 min 90% A → 2.5 min % A → 3.0 min 5% A → 4.5 min 5% A; flow rate: 0.0 min 1 ml/min → 2.5 min/3.0 min/4.5 min 2 ml/min; oven: 50°C; UV detection: 210 nm.
Method 5 (HPLC): Instrument: HP 1090 Series II; column: Merck Chromolith SpeedROD RP-18e, 50 mm x 4.6 mm; preliminary column: Merck Chromolith Guard Cartridge Kit RP-18e, 5 mm x 4.6 mm; injection volume: 5 µl; eluent A: 70% HClO4 in water (4 ml/litre), eluent B: acetonitrile; gradient: 0.00 min % B → 0.50 min 20% B → 3.00 min 90% B → 3.50 min 90% B → 3.51 min 20% B → 4.00 min 20% B; flow rate: 5 ml/min; column temperature: 40°C.
Method 6 (HPLC): Instrument: Waters 2695 with DAD 996; column: Merck Chromolith SpeedROD RP-18e, 50 mm x 4.6 mm; Ord. No.: 1.51450.0001, inary column: Merck Chromolith Guard Cartridge Kit RP- 18e, 5 mm x 4.6 mm; Ord. No.: 0.0001, eluent A: 70% HClO4 in water (4 ml/litre), eluent B: acetonitrile; gradient: 0.00 min 5% B → 0.50 min 5% B → 3.00 min 95% B → 4.00 min 95% B; flow rate: 5 ml/min.
Method 7 (LC-MS): MS instrument: Waters ZQ; HPLC instrument: Agilent 1100 ; UV DAD; column: Thermo Hypersil GOLD 3µ 20 mm x 4 mm; eluent A: 1 l water + 0.5 ml 50% strength formic acid, eluent B: 1 l acetonitrile + 0.5 ml 50% strength formic acid; gradient: 0.0 min 100% A → 3.0 min 10% A → 4.0 min 10% A → 4.1 min 100% A (flow rate 2.5 ml/min); oven: 55°C; flow rate: 2 ml/min; UV detection: 210 nm.
Method 8 (LC-MS): MS instrument: Waters ZQ; HPLC instrument: Agilent 1100 Series; UV DAD; column: Thermo Hypersil GOLD 3µ 20 mm x 4 mm; eluent A: 1 l water + 0.5 ml 50% strength formic acid, eluent B: 1 l acetonitrile + 0.5 ml 50% th formic acid; gradient: 0.0 min 100% A → 2.0 min 60% A → 2.3 min 40% A → 3.0 min 20% A → 4.0 min 10% A → 4.2 min 100% A (flow rate 2.5 ml/min); oven: 55°C; flow rate: 2 ml/min; UV detection: 210 nm.
Method 9 (LC-MS): Instrument: Waters Acquity SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8µ 50 mm x 1 mm; eluent A: 1 l water + 0.25 ml 99% strength formic acid, eluent B: 1 l acetonitrile + 0.25 ml 99% strength formic acid; nt: 0.0 min 95% A → 6.0 min 5% A → 7.5 min 5% A; oven: 50°C; flow rate: 0.35 ml/min; UV detection: 210-400 nm.
Method 10 : Instrument: Agilent 1200 Series; column: Agilent Eclipse 8 5µ 4.6 mm x 150 mm; preliminary column: Phenomenex KrudKatcher Disposable Pre-Column; injection volume: 5 µl; eluent A: 1 l water + 0.01% trifluoroacetic acid; eluent B: 1 l acetonitrile + 0.01% trifluoroacetic acid; nt: 0.00 min 10% B → 1.00 min 10% B → 1.50 min 90% B → 5.5 min 10% B; flow rate: 2 ml/min; column temperature: 30°C.
For all reactants or reagents whose preparation is not explicitly described below, they were ed commercially from generally available sources. For all other reactants or reacents whose preparation is likewise not described below, and which were not available commercially or were obtained from sources which are not generally available, a reference is given to the published literature in which their preparation is described.
Method 11 (LC-MS): Instrument: Waters ACQUITY SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8 µ x 2 mm; eluent A: 1 l water + 0.25 ml 99% strength formic acid , eluent B: 1 l acetonitrile + 0.25 ml 99% strength formic acid; gradient: 0.0 min 90% A → 1.2 min 5% A → 2.0 min 5% A oven: 50°C; flow rate: 0.60 ; UV ion: 208 – 400 nm.
Method 12 (HPLC): ment: Agilent 1200 Series with column oven and DAD; column: Merck Chromolith SpeedROD RP-18e, 50 mm x 4.6 mm; Ord. No.: 1.51450.0001; preliminary column: Merck Chromolith Guard Cartridge Kit RP-18e, 5 mm x 4.6 mm; Ord. No.: 0.0001; eluent A: 70% HClO 4 in water (4 ml/litre), eluent B: acetonitrile; gradient: 0.00 min 5% B → 0.50 min 5% B → 3.00 min 95% B → 4.00 min 95% B; flow rate: 5 ml/min; column temperature: 30°C.
Starting Compounds and Intermediates: Starting nd 1 (2R,3R)[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoic acid (Bocdolaproine H C CH 3 N O O O O CH 3 The title compound can be prepared in various ways according to literature methods; see, for example, Pettit et al., Synthesis 1996, 719; Shioiri et al., Tetrahedron Lett. 1991, 32, 931; Shioiri et al., Tetrahedron 1993, 49, 1913; Koga et al., Tetrahedron Lett. 1991, 32, 2395; Vidal et al., Tetrahedron 2004, 60, 9715; Poncet et al., edron 1994, 50, 5345. It was prepared either as the free acid or as a 1:1 salt with dicyclohexylamine.
Starting nd 2a tert-butyl (3R,4S,5S)methoxymethyl(methylamino)heptanoate hydrochloride soleucine-OtBu x HCl) H C H C 3 3 3 CH H C O 3 N H x HCl O O The title compound can be prepared in various ways according to literature methods; see, for example, Pettit et al., J. Org. Chem. 1994, 59, 1796; Koga et al., Tetrahedron Lett. 1991, 32, 2395; Shioiri et al., Tetrahedron Lett. 1991, 32, 931; Shioiri et al., Tetrahedron 1993, 49, 1913.
Starting Compound 2b tert-butyl (3R,4S,5S)methoxymethyl(methylamino)heptanoate (dolaisoleucine-OtBu) H C H C 3 3 3 CH H C O 3 N O O The compound was prepared in analogy to starting compound 2a, except that the hydrogenation was performed without addition of 1N hydrochloric acid. ng nd 3 Nα-(tert-butoxycarbonyl)-N-hydroxy-L-phenylalaninamide H C H 3 O N H3C O CH O The title compound was prepared by the literature method (A. Ritter et al., J. Org. Chem. 1994, 59, 4602).
Yield: 750 mg (75% of theory) LC-MS (Method 3): Rt = 1.67 min; MS (ESIpos): m/z = 281 (M+H)+.
Starting Compound 4 1,2-oxazolidine hydrochloride x HCl The title compound can be prepared by literature methods (see, for example, H. King, J. Chem.
Soc. 1942, 432); it is also commercially available. ng Compound 5 1,2-oxazinane hydrochloride O x HCl The title compound can be prepared by literature methods (see, for example, H. King, J. Chem.
Soc. 1942, 432).
Starting Compound 6 2-oxaazabicyclo[2.2.2]octene The title compound can be prepared in otected form by the literature method (see, for example, C. Johnson et al., edron Lett. 1998, 39, 2059); the deprotection was effected in a customary manner by treatment with trifluoroacetic acid and subsequent neutralization.
Yield: 149 mg (89% of theory) Starting Compound 7 tert-butyl (1S,2R)(hydroxycarbamoyl)phenylcyclopropyl carbamate H C H 3 O N H C O CH O The title compound was prepared by a literature method (A. Ritter et al., J. Org. Chem. 1994, 59, 4602) proceeding from commercially available (1S,2R)[(tert-butoxycarbonyl)amino] phenylcyclopropanecarboxylic acid (C. ela et al., Chirality 1999, 11, 583).
Yield: 339 mg (59% of theory) LC-MS (Method 1): Rt = 0.82 min; MS s): m/z = 293 (M+H)+.
Intermediate 1 tert-butyl ,5S)[{N-[(benzyloxy)carbonyl]-L-valyl}(methyl)amino]methoxy methylheptanoate H3C H C 3 O CH3 O N O O CH O O H C CH CH3 3 3 10.65 g (41.058 mmol) of utyl (3R,4S,5S)methoxymethyl(methylamino)heptanoate (starting nd 2b) were taken up in 250 ml of dichloromethane and the solution was cooled to -10°C. Then, while stirring, 10.317 g (41.058 mmol) of N-[(benzyloxy)carbonyl]-L-valine, 16.866 g (61.586 mmol) of 2-bromoethylpyridinium tetrafluoroborate (BEP) and 28.6 ml of N,N-diisopropylethylamine were added, and the mixture was subsequently stirred at RT for 20 h.
The reaction mixture was then diluted with dichloromethane and shaken twice with saturated sodium chloride on, dried over sodium sulphate, filtered and concentrated. The residue was purified by flash chromatography on silica gel with 4:1 petroleum ether/ethyl acetate as the eluent.
The ponding ons were concentrated and the residue was dried under high vacuum overnight. 10.22 g (51% of theory) of the title compound were obtained as a yellowish oil.
HPLC (Method 5): Rt = 2.3 min; LC-MS (Method 2): Rt = 1.59 min; MS (ESIpos): m/z = 493 (M+H)+.
Intermediate 2 tert-butyl (3R,4S,5S)methoxymethyl[methyl(L-valyl)amino]heptanoate H C 3 3 H C O 3 CH H N O 2 N CH O O H C CH 3 CH 3 3 3 500 mg (1 mmol) of tert-butyl (3R,4S,5S)[{N-[(benzyloxy)carbonyl]-L-valyl}(methyl)amino] methoxymethylheptanoate (intermediate 1) were dissolved in 50 ml of methanol and, after addition of 100 mg of 10% palladium on activated carbon, hydrogenated under standard hydrogen re at RT for 1 h. The catalyst was then filtered off and the solvent was removed under reduced pressure. This gave 370 mg (quant.) of the title compound as a lly colourless oil.
HPLC (Method 5): Rt = 1.59 min; LC-MS (Method 1): Rt = 0.74 min; MS (ESIpos): m/z = 359 (M+H)+. ediate 3 N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(3R,4S,5S)tert-butoxymethoxy- -methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C H C 3 3 3 3 O 3 O CH N O O N N CH O CH O O 3 3 H C CH CH 3 3 3 4.64 g (13.13 mmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valine were dissolved in 20 ml of DMF and admixed successively with 4.28 g (11.94 mmol) of tert-butyl (3R,4S,5S) methoxymethyl[methyl(L-valyl)amino]heptanoate (Intermediate 2), 2.75 g (14.33 mmol) of 1-(3-dimethylaminopropyl)ethylcarbodiimide hydrochloride and 2.2 g (14.33 mmol) of 1- hydroxy-1H-benzotriazole hydrate. The mixture was stirred at RT overnight. The reaction mixture was then poured into a mixture of semisaturated aqueous um chloride solution and ethyl acetate. The organic phase was removed, washed sively with saturated sodium encarbonate solution and saturated sodium chloride solution, dried over magnesium sulphate, filtered and concentrated. The residue was used directly in the next stage, without further purification.
Yield: 9.1 g (quant., 60% purity) HPLC d 5): Rt = 2.7 min; LC-MS (Method 2): Rt = 1.99 min; MS (ESIpos): m/z = 694 (M+H)+.
Intermediate 4 N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)carboxymethoxy methylhexanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 O O N OH O N N CH O CH O O 3 3 H C CH CH 3 3 3 9.1 g of the crude product N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(3R,4S,5S)- 1-tert-butoxymethoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 3) were taken up in 56.6 ml of dichloromethane, 56.6 ml of trifluoroacetic acid were added, and the mixture was stirred at RT for 2 h. Subsequently, the reaction mixture was concentrated under reduced re and the remaining residue was purified by flash chromatography, using dichloromethane, 3:1 dichloromethane/ethyl acetate and 15:5:0.5 dichloromethane/ethyl acetate/methanol as eluent. After purification of the corresponding fractions and concentration, 5.8 g (86% of ) of the title compound were ed as a colourless foam.
HPLC (Method 5): Rt = 2.2 min; LC-MS (Method 1): Rt = 1.3 min; MS (ESIpos): m/z = 638 (M+H)+.
Intermediate 5 tert-butyl (2S)(1,2-oxazinanyl)oxophenylpropanyl carbamate 3 CH3 O H C N 3 H O N 500 mg (1.9 mmol) of N-(tert-butoxycarbonyl)-L-phenylalanine were ved in 10 ml of DMF and admixed successively with 466 mg (3.8 mmol) of 1,2-oxazinane hydrochloride (Starting Compound 5), 433 mg (2.3 mmol) of imethylaminopropyl)ethylcarbodiimide hydrochloride, 382 mg (2.8 mmol) of 1-hydroxy-1H-benzotriazole hydrate and 731 mg (5.7 mmol) of isopropylethylamine. The mixture was stirred at RT overnight. The reaction mixture was then poured into a mixture of semisaturated aqueous ammonium chloride solution and ethyl e. The organic phase was removed, washed sively with saturated sodium hydrogencarbonate solution and saturated sodium chloride solution, dried over magnesium sulphate, filtered and concentrated. 620 mg (98% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 2): Rt = 1.62 min; MS (ESIpos): m/z = 235 (M-C4H8-CO2+H)+.
Intermediate 6 -amino(1,2-oxazinanyl)phenylpropanone trifluoroacetate CF3COOH x H N 2 O 620 mg (1.85 mmol) of tert-butyl (2S)(1,2-oxazinanyl)oxophenylpropanyl carbamate (Intermediate 5) were taken up in 5 ml of romethane, 10 ml of trifluoroacetic acid were added and the mixture was stirred at RT for 30 min. Subsequently, the reaction e was concentrated under reduced pressure and the remaining residue was lyophilized from water/acetonitrile. In this way, 779 mg (91% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 0.45 min; LC-MS (Method 3): Rt = 1.09 min; MS (ESIpos): m/z = 235 (M+H)+.
Intermediate 7 (2R,3R)methoxymethyl-N-[(2S)(1,2-oxazinanyl)oxophenylpropanyl][(2S)- pyrrolidinyl]propanamide trifluoroacetate WO 87716 3 N CF3COOH x N O O O 360 mg (1.25 mmol) of (2R,3R)[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxy methylpropanoic acid ing Compound 1) were taken up in 10 ml of DMF and admixed successively with 579.2 mg (1.25 mmol) of (2S)amino(1,2-oxazinanyl)phenylpropan one trifluoroacetate (Intermediate 6), 714.5 mg (1.88 mmol) of O-(7-azabenzotriazolyl)- N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 655 µl of N,N- diisopropylethylamine. The mixture was stirred at RT for 16 h. The reaction mixture was then concentrated, and the residue was taken up in ethyl acetate and extracted by shaking first with 5% aqueous citric acid solution, then with 5% aqueous sodium hydrogencarbonate solution and subsequently with saturated sodium chloride solution. The c phase was concentrated and the residue was ed by flash chromatography on silica gel with 16:4 dichloromethane/methanol as the . The corresponding fractions were combined and the solvent was removed under reduced pressure. After the residue had been dried under high vacuum, 503.5 mg (74% of theory) of the Boc-protected intermediate utyl (2S)[(1R,2R)methoxymethyl{[(2S)(1,2- oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidinecarboxylate were obtained.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 1.12 min; MS (ESIpos): m/z = 504 (M+H)+. 503 mg (1 mmol) of this intermediate were taken up in 20 ml of dichloromethane, 10 ml of trifluoroacetic acid were added, and the mixture was stirred at RT for 30 min. Subsequently, the reaction mixture was concentrated under reduced re and redistilled with dichloromethane.
The remaining residue was precipitated from ethyl acetate with n-pentane, and the solvent was decanted off. The residue thus obtained was ved in water and extracted by shaking with ethyl acetate, and the aqueous phase was subsequently lyophilized. In this way, 462 mg (89% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 12): Rt = 1.53 min; LC-MS d 11): Rt = 0.57 min; MS (ESIpos): m/z = 404 (M+H)+.
Intermediate 8 N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(2R,3S,4S)carboxymethoxymethylhexan yl]-N-methyl-L-valinamide H C CH3 H3C CH3 O O N OH 3 O N N 3 CH O CH O O 3 3 H C CH CH3 3 3 51 mg (0.08 mmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S) carboxymethoxymethylhexanyl]-N-methyl-L-valinamide mediate 4) were dissolved in 10 ml of DMF, and 0.5 ml of piperidine was added. After stirring at RT for 10 min, the reaction mixture was concentrated under reduced pressure and the residue was stirred with diethyl ether.
The insoluble constituents were ed off and washed repeatedly with diethyl ether. Then the filter residue was taken up in 5 ml of dioxane/water and the solution was adjusted to pH 11 with 1 N sodium hydroxide solution. Under ultrasound treatment, a total of 349 mg (1.6 mmol) of di-tertbutyl dicarbonate were added in several ns, in the course of which the pH of the solution was kept at 11. After the reaction had ended, the dioxane was evaporated off and the aqueous solution was adjusted to a pH of 2-3 with citric acid. The mixture was extracted twice with 50 ml each time of ethyl acetate. The organic phases were combined, dried over magnesium sulphate and concentrated under reduced re. The residue was taken up in diethyl ether and the of the title nd was precipitated with pentane. The solvent was removed by decantation. The residue was digested several times more with pentane and finally dried under high vacuum. 40 mg (97% of ) of the title compound were thus obtained.
HPLC d 6): Rt = 2.2 min; LC-MS (Method 2): Rt = 1.32 min; MS (ESIpos): m/z = 516 (M+H)+.
Intermediate 9 tert-butyl (2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl) phenylcyclopropyl]amino}oxopropyl]pyrrolidinecarboxylate 3 N H C CH 3 N O 3 O O O O CH The title compound was prepared in analogy to the synthesis of Intermediates 5, 6 and 7 over three stages, by coupling of commercially available (1S,2R)[(tert-butoxycarbonyl)amino] phenylcyclopropanecarboxylic acid with 1,2-oxazinane hydrochloride (Starting Compound 5), uent deprotection with trifluoroacetic acid and ng with Starting Compound 1. The end product was purified by preparative HPLC.
HPLC (Method 5): Rt = 2.12 min; LC-MS d 2): Rt = 1.25 min; MS (ESIpos): m/z = 516 (M+H)+.
Intermediate 10 N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide H C CH H C CH 3 3 3 3 O O H OH N N O N N O O CH O CH O O CH 3 3 3 H C CH CH 3 3 3 315 mg (0.494 mmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)- oxymethoxymethylhexanyl]-N-methyl-L-valinamide (Intermediate 4) were dissolved in 12 ml of DMF, and admixed with 104 mg (0.543 mmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride and 83 mg (0.543 mmol) of 1-hydroxy- 1H-benzotriazole hydrate, and the mixture was stirred at RT for 90 min. Subsequently, 112 µl of N,N-diisopropylethylamine and 149 mg (0.494 mmol) of (2 R,3R)methoxymethyl[(2S)- pyrrolidinyl]propanoic acid trifluoroacetate, which had been prepared beforehand from Starting Compound 1 by elimination of the Boc protecting group by means of oroacetic acid, were added. The mixture was stirred at RT for 2 h and then concentrated under high . The remaining residue was purified twice by preparative HPLC. 140 mg (35% of theory) of the title compound were obtained in the form of a colourless foam.
HPLC (Method 5): Rt = 2.40 min; LC-MS (Method 1): Rt = 1.38 min; MS s): m/z = 807 (M+H)+.
Intermediate 11 N-[(benzyloxy)carbonyl]-N-methyl-L-threonyl-N-[(2R,3S,4S)carboxymethoxy methylhexanyl]-N-methyl-L-valinamide H C OH H C 3 3 O O N OH O N N CH O CH O O 3 3 H C CH CH 3 3 3 First, N-[(benzyloxy)carbonyl]-N-methyl-L-threonine was released from 237 mg (0.887 mmol) of its dicyclohexylamine salt thereof by taking it up in ethyl acetate and extractive shaking with 5% aqueous sulphuric acid. The organic phase was dried over magnesium sulphate, filtered and concentrated. The e was taken up in 16 ml of DMF and admixed successively with 365 mg (1 mmol) of tert-butyl ,5S)methoxymethyl[methyl(L-valyl)amino]heptanoate (Intermediate 2), 185 mg (0.967 mmol) of 1-(3-dimethylaminopropyl)ethylcarbodiimide hydrochloride and 148 mg (0.967 mmol) of 1-hydroxy-1H-benzotriazole hydrate. The mixture was stirred at RT for 2 h. The reaction mixture was then poured into a mixture of semisaturated aqueous ammonium chloride solution and ethyl e. The organic phase was removed, washed successively with saturated sodium hydrogencarbonate solution and saturated sodium chloride solution, dried over magnesium sulphate, filtered and concentrated. The residue was purified by preparative HPLC. 283 mg (53% of theory) of the utyl ester intermediate N- [(benzyloxy)carbonyl]-N-methyl-L-threonyl-N-[(3R,4S,5S)tert-butoxymethoxymethyl oxoheptanyl]-N-methyl-L-valinamide were thus obtained.
HPLC (Method 5): Rt = 2.17 min. 283 mg (0.466 mmol) of this intermediate were taken up in 5 ml of dichloromethane, 5 ml of ous trifluoroacetic acid were added, and the mixture was d at RT for 2 h.
Subsequently, the reaction mixture was concentrated under high vacuum and the remaining residue was purified by means of preparative HPLC. This gave 156 mg (61% of theory) of the title nd as a colourless foam.
HPLC (Method 5): Rt = 1.50 min; LC-MS (Method 2): Rt = 1.09 min; MS (ESIpos): m/z = 552 (M+H)+.
Intermediate 12 benzyl N-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-L-phenylalaninate oroacetate CH O 3 H CF3COOH x N O O O In the first step, Starting Compound 1 was released from 600 mg (1.28 mmol) of the corresponding dicyclohexylammonium salt by dissolving the salt in 100 ml of ethyl acetate and extractive shaking, first with 50 ml of 0.5% sulphuric acid and then with saturated sodium de solution.
Then the organic phase was dried over magnesium sulphate, filtered, concentrated and d immediately with benzyl L-phenylalaninate in analogy to the synthesis of Intermediate 7, and then deprotected.
Yield: 650 mg (94% over 2 stages) HPLC (Method 6): Rt = 1.76 min; LC-MS (Method 2): Rt = 1.68 min; MS (ESIpos): m/z = 425 (M+H)+.
Intermediate 13 benzyl (βS)-N-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-β-methyl-L- phenylalaninate trifluoroacetate CH O 3 H CF3COOH x N O O O 3 H3C First, (2R,3R)[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoic acid was released from 351 mg (0.75 mmol) of the dicyclohexylamine salt (Starting Compound 1) by taking it up in ethyl acetate and tive shaking with aqueous 5% potassium hydrogensulphate solution. The organic phase was dried over ium sulphate, filtered and concentrated. The residue was taken up in 10 ml of DMF and admixed successively with 373 mg (0.75 mmol) of benzyl (βS)-β-methyl-L-phenylalaninate trifluoroacetate [prepared from commercially available (βS)-N-(tert-butoxycarbonyl)-β-methyl-L-phenylalanine by EDC/DMAP-mediated esterification with benzyl alcohol and uent detachment of the Boc protecting group with trifluoroacetic acid], 428 mg (1.125 mmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 392 µl of N,N-diisopropylethylamine. The mixture was stirred at RT for 20 h. The reaction mixture was then poured onto a mixture of turated aqueous ammonium de solution and ethyl acetate. The organic phase was removed, washed successively with saturated sodium hydrogencarbonate solution and saturated sodium de solution, and subsequently concentrated. The residue was purified by means of preparative HPLC.
This gave 230 mg (57% of theory) of the Boc-protected ediate benzyl (βS)-N-{(2R,3R) [(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoyl}-β-methyl-L- phenylalaninate.
HPLC (Method 6): Rt = 2.3 min; LC-MS (Method 1): Rt = 1.36 min; MS (ESIpos): m/z = 539 (M+H)+. 230 mg (0.42 mmol) of this intermediate were taken up in 5 ml of dichloromethane, 5 ml of trifluoroacetic acid were added, and the mixture was stirred at RT for 30 min. Subsequently, the reaction mixture was concentrated under reduced pressure. The remaining residue was the on mixture dried further under d pressure and then lyophilized from acetonitrile/water. In this way, 230 mg (quant.) of the title compound were obtained.
HPLC (Method 6): Rt = 1.6 min.
Intermediate 14 N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S) (1,2-oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C CH N 3 3 3 O 3 H H N N N O HN N O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 x CF3COOH 143 mg (0.223 mmol) of -fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)- 1-carboxymethoxymethylhexanyl]-N-methyl-L-valinamide (Intermediate 4) were taken up in 15 ml of DMF and d successively with 141 mg (0.22 mmol) of (2R,3R)methoxy methyl-N-[(2S)(1,2-oxazinanyl)oxophenylpropanyl][(2S)-pyrrolidin yl]propanamide trifluoroacetate (Intermediate 7), 102 mg (0.27 mmol) of O-(7-azabenzotriazol yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 128 µl (0.74 mmol) of N,N- diisopropylethylamine. The mixture was stirred at RT for 3 h. The reaction mixture was then poured into a mixture of semisaturated aqueous ammonium chloride solution and ethyl acetate.
The organic phase was removed, washed successively with saturated sodium hydrogencarbonate solution and saturated sodium chloride solution, dried over magnesium te, filtered and concentrated. This gave 275 mg (quant.) of the Fmoc-protected intermediate N-[(9H-fluoren ylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy- 2-methyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide.
HPLC d 5): Rt = 2.73 min; LC-MS (Method 4): Rt = 3.19 min; MS (ESIpos): m/z = 1023 (M+H)+. 46 mg (0.045 mmol) of this intermediate were dissolved in 4 ml of DMF. After 1 ml of piperidine had been added, the on mixture was stirred at RT for 1 h. Subsequently, the reaction e was concentrated under d pressure and the residue was purified by means of preparative HPLC (eluent: acetonitrile + 0.01% TFA / water + 0.01% TFA). 22 mg (54% of theory) of the title compound were obtained as a colourless foam.
WO 87716 HPLC (Method 5): Rt = 1.68 min; LC-MS (Method 2): Rt = 1.03 min; MS (ESIpos): m/z = 801 (M+H)+ 1H NMR (600 MHz, DMSO-d 6): δ = 8.8 (m, 2H), 8.7 (m, 1H), 8.42 and 8.15 (2d, 1H), 7.3-7.1 (m, 5H), 5.12 and 4.95 (2m, 1H), 4.70 and 4.62 (2m, 1H), 4.62 and 4.50 (2t, 1H), 4.1-3.9 (m, 3H), 3.85 (m, 1H), 3.75-3.6 (m, 2H), 3.23, 3.18, 3.17, 3.14, 3.02 and 2.96 (6s, 9H), 3.1-2.9 and 2.75 (2m, 2H), 2.46 (m, 3H), 2.4-2.1 (m, 2H), 2.05 (br. m, 2H), 1.85-1.55 (br. m, 6H), 1.5-1.2 (br. m, 3H), 8 (m, 18H), 0.75 (t, 3H) [further s hidden under H2O peak].
Intermediate 15 N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S,3S)- 1-(1,2-oxazinanyl)oxophenylbutanyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C CH3 N 3 3 3 O H H N N N O HN N O O CH O CH3 O O H C 3 3 H C H C CH CH3 3 3 3 x CF3COOH 126 mg (0.198 mmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)- 1-carboxymethoxymethylhexanyl]-N-methyl-L-valinamide (Intermediate 4) were taken up in 10 ml of DMF and admixed successively with 105 mg (0.198 mmol) of (2R,3R)methoxy methyl-N-[(2S,3S)(1,2-oxazinanyl)oxophenylbutanyl][(2S)-pyrrolidin yl]propanamide trifluoroacetate (Intermediate 17), 41.6 mg (0.217 mmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 33 mg (0.217 mmol) of 1-hydroxy-1H- benzotriazole hydrate and 79 µl (0.454 mmol) of isopropylethylamine. The mixture was stirred at RT overnight. The reaction mixture was then poured into a mixture of semisaturated aqueous ammonium chloride solution and ethyl acetate. The organic phase was removed, washed successively with ted sodium hydrogencarbonate solution and saturated sodium de solution, dried over magnesium sulphate, filtered and concentrated. This gave 220 mg (quant.) of the Fmoc-protected intermediate N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N- [(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S,3S)(1,2-oxazinanyl)- 2012/075277 1-oxophenylbutanyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N- methyl-L-valinamide.
HPLC d 5): Rt = 2.77 min; LC-MS (Method 1): Rt = 1.5 min; MS (ESIpos): m/z = 1037 . 220 mg (0.212 mmol) of this intermediate were dissolved in 5 ml of DMF. After 1 ml of piperidine had been added, the reaction mixture was stirred at RT for 1 h. Subsequently, the reaction mixture was concentrated under reduced pressure and the residue was purified by means of preparative HPLC (eluent: acetonitrile + 0.01% TFA / water + 0.01% TFA). 91 mg (46% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.71 min; LC-MS d 1): Rt = 0.9 min; MS (ESIpos): m/z = 815 (M+H)+ 1H NMR (600 MHz, DMSO-d 6): δ = 8.87 and 8.80 (2d, 2H), 8.75 (m, 1H), 8.40 and 7.98 (2d, 1H), 7.3-7.1 (m, 5H), 5.45 and 5.2 (2t, 1H), 4.78 and 4.62 (2m, 1H), 4.73 and 4.58 (2t, 1H), 4.2-4.0 (m, 3H), 3.7-3.6 (m, 1H), 3.35, 3.20, 3.18, 3.14, 3.12 and 3.00 (6s, 9H), 3.1 and 2.95 (2m, 2H), 2.46 (m, 3H), 2.4-2.0 (m, 4H), 1.9-1.6 (m, 4H), 1.6-1.2 (m, 5H), 1.1-0.75 (m, 21H), 0.80 (t, 3H) [further signals hidden under H2O peak].
Intermediate 16 N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R)- 1-(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl} methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C 3 3 CH3 3 N O H H N N N O HN N O O CH O CH O O H C 3 3 3 H C CH CH3 3 3 617 mg (1.2 mmol) of tert-butyl (2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinan- 2-ylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinecarboxylate (Intermediate 24) were taken up in 44 ml of dichloromethane, 4.4 ml of trifluoroacetic acid were added and the e was stirred at RT for 30 min. Subsequently, the reaction mixture was concentrated under reduced pressure and the remaining residue was lyophilized from dioxane/water. 702 mg (quant.) of the deprotected compound (2R,3 ethoxymethyl-N-[(1 S,2 1,2-oxazinan ylcarbonyl)phenylcyclopropyl][(2 S)-pyrrolidinyl]propanamide trifluoroacetate were obtained as a crude product, which was used in the following stage without further cation. 470 mg (0.74 mmol) of N-[(9 H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2 R,3 S,4 S) carboxymethoxymethylhexanyl]-N-methyl-L-valinamide (Intermediate 4) were taken up in 57 ml of DMF and admixed successively with 390 mg (approx. 0.74 mmol) of the above-obtained (2 R,3 R)methoxymethyl-N-[(1 S,2 R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] [(2 S)-pyrrolidinyl]propanamide trifluoroacetate, 336 mg (0.88 mmol) of zabenzotriazol- N,N,N',N' -tetramethyluronium hexafluorophosphate (HATU) and 423 µl (2.4 mmol) of N,N - diisopropylethylamine. The mixture was d at RT for 2 h. The on mixture was then poured into a mixture of semisaturated aqueous ammonium chloride solution and ethyl acetate.
The organic phase was removed, washed successively with saturated sodium hydrogencarbonate solution and saturated sodium chloride solution, dried over sodium sulphate, filtered and concentrated. The e was purified by preparative HPLC. This gave 453 mg (59% of theory) of the Fmoc-protected intermediate N-[(9 H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N- [(3 R,4 S,5 S)methoxy{(2 S)[(1 R,2 R)methoxymethyl{[(1 S,2 R)(1,2-oxazinan ylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptan yl]-N-methyl-L-valinamide.
HPLC (Method 5): Rt = 2.58 min; LC-MS (Method 1): Rt = 3.10 min; MS (ESIpos): m/z = 1035 (M+H)+. 453 mg (0.438 mmol) of this intermediate were dissolved in 24 ml of DMF. After 2.4 ml of piperidine had been added, the reaction mixture was stirred at RT for 30 min. Subsequently, the reaction mixture was concentrated under reduced pressure and the residue was purified by preparative HPLC (eluent: acetonitrile / 0.1% TFA in water). 260 mg (64% of ) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.64 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 813 (M+H)+ 1H NMR (400 MHz, DMSO-d 6): δ = 8.8 (m, 2H), 8.65 (m, 2H), 1 (m, 5H), 4.8-4.05 (m, 2H), 4.0 and 3.82 (2m, 2H), 3.8-3.5 (m, 8H), 3.32, 3.29, 3.20, 3.19, 3.12 and 3.00 (6s, 9H), 2.65 (t, 1H), 45 (m, 3H), 3 (m, 15H), 1.15-0.85 (m, 18H), 0.8 and 0.75 (2d, 3H) [further signals hidden under H2O peak].
Intermediate 17 N-benzyl-N-methyl-L-phenylalaninamide trifluoroacetate 3 N CF3COOH x H N 2 O 1000 mg (3.77 mmol) of N-(tert-butoxycarbonyl)-L-phenylalanine were dissolved in 10 ml of DMF and admixed with 457 mg (3.77 mmol) of N-methylbenzylamine, 2150 mg (5.65 mmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 657 µl of N,N- ropylethylamine. The reaction mixture was stirred at RT for 30 min and then concentrated under reduced pressure. The residue was taken up in dichloromethane and extracted by shaking three times with water. The organic phase was dried over magnesium sulphate and trated.
The residue was purified by flash chromatography on silica gel with 3:1 petroleum ether/ethyl acetate as the eluent. The product fractions were concentrated and the e was dried under high vacuum. This gave 1110 mg (75% of theory) of the Boc-protected intermediate N-benzyl-Nα-(tert- butoxycarbonyl)-N-methyl-L-phenylalaninamide.
HPLC (Method 6): Rt = 2.1 min; LC-MS (Method 1): Rt = 1.14 min; MS (ESIpos): m/z = 369 (M+H)+. 1108 mg (3,007 mmol) of this intermediate were taken up in 30 ml of dichloromethane, 10 ml of trifluoroacetic acid were added, and the mixture was stirred at RT for 30 min. Subsequently, the reaction e was concentrated under reduced pressure, the remaining residue was stirred with dichloromethane and the solvent was distilled off. The e was stirred twice more with pentane, the solvent was decanted off again each time and the of the title compound was finally dried under high vacuum. 1075 mg (93% of theory) of the title compound were thus obtained as a resin.
HPLC (Method 6): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.6 min; MS (ESIpos): m/z = 269 (M+H)+.
Intermediate 18 N-benzyl-Nα-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-N-methyl-L- phenylalaninamide trifluoroacetate CH 3 N 3 H CF3COOH x N O O O First, (2R,3R)[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoic acid (Starting Compound 1) was ed from 141 mg (0.491 mmol) of its dicyclohexylamine salt by taking it up in ethyl acetate and tive shaking with 5% aqueous sulphuric acid. The organic phase was dried over magnesium sulphate, filtered and concentrated. The residue was taken up in ml of DMF and 187.6 mg (0.49 mmol) of N-benzyl-N-methyl-L-phenylalaninamide trifluoroacetate (Intermediate 9), 190.3 mg (1.47 mmol) of O-(7-azabenzotriazolyl)-N,N,N',N'- tetramethyluronium hexafluorophosphate (HATU) and 256 µl of N,N-diisopropylethylamine were added. The mixture was stirred at RT for 1 h. The reaction mixture was then concentrated, the residue was taken up in ethyl acetate and the solution was subsequently extracted by shaking successively with saturated ammonium chloride solution, saturated sodium hydrogencarbonate on and water. The organic phase was dried over ium sulphate and concentrated. The residue was ed by flash chromatography on silica gel with 30:1 acetonitrile/water as the . The product fractions were concentrated and the residue was dried under high vacuum.
This gave 168 mg (64% of theory) of the Boc-protected intermediate tert-butyl (2S)[(1R,2R) ({(2S)[benzyl(methyl)amino]oxophenylpropanyl}amino)methoxymethyl oxopropyl]pyrrolidinecarboxylate.
HPLC (Method 6): Rt = 2.2 min; LC-MS (Method 2): Rt = 1.22 min; MS (ESIpos): m/z = 538 (M+H)+. 168 mg (0.312 mmol) of this intermediate were taken up in 15 ml of dichloromethane, 3 ml of trifluoroacetic acid were added, and the mixture was stirred at RT for 30 min. Subsequently, the reaction mixture was trated under reduced pressure. The remaining e was stirred first with dichloromethane, then with diethyl ether, and the solvent was distilled off again each time.
After drying under high vacuum, 170 mg (99% of theory) of the title compound were obtained as a resin.
HPLC (Method 6): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.73 min; MS (ESIpos): m/z = 438 (M+H)+.
Intermediate 19 methyl N-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-L-phenylalaninate trifluoroacetate CH O 3 3 H CF3COOH x N O O O The title nd was prepared in analogy to the synthesis of Intermediate 18, proceeding from (2R,3R)[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoic acid (Starting Compound 1), which was released from the dicyclohexylamine salt, and methyl L- phenylalaninate hydrochloride.
HPLC (Method 5): Rt = 0.6 min; LC-MS (Method 3): Rt = 1.17 min; MS (ESIpos): m/z = 349 .
Intermediate 20 benzyl N-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-L-tryptophanate trifluoroacetate CH O 3 H CF3COOH x N O O O The title compound was prepared in analogy to the synthesis of Intermediate 18, proceeding from )[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoic acid (Starting Compound 1), which was released from the ohexylamine salt, and benzyl L- tryptophanate.
WO 87716 HPLC (Method 6): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.8 min; MS (ESIpos): m/z = 464 (M+H)+.
Intermediate 21 benzyl (1S,2R)({(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}amino) phenylcyclopropanecarboxylate trifluoroacetate 3 O CF3COOH x N O O O The title compound was prepared in analogy to the synthesis of Intermediate 18, proceeding from (2R,3R)[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoic acid (Starting Compound 1), which was released from the dicyclohexylamine salt, and benzyl (1S,2R)- ophenylcyclopropanecarboxylate. Benzyl (1S,2R)amino phenylcyclopropanecarboxylate had been prepared beforehand by standard methods, by esterifying commercially available (1S,2R)[(tert-butoxycarbonyl)amino]phenylcyclopropanecarboxylic acid with benzyl alcohol and subsequent Boc detachment with trifluoroacetic acid.
HPLC d 5): Rt = 1.5 min; LC-MS d 2): Rt = 0.93 min; MS (ESIpos): m/z = 437 (M+H)+.
Intermediate 22 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-N'-methylhexanehydrazide trifluoroacetate O CH CF3COOH x 3 N NH 100 mg (473 µmol) of 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoic acid were dissolved in 71 µl of DMF and then admixed with 139 mg (947 µmol) of tert-butyl ylhydrazinecarboxylate, 182 mg (947 µmol) of 1-(3-dimethylaminopropyl)ethylcarbodiimide hydrochloride and 145 mg (947 µmol) of 1-hydroxy-1H-benzotriazole hydrate. The mixture was stirred at RT overnight and then concentrated under reduced pressure. The remaining residue was purified by means of preparative HPLC. After lization from dioxane/water, 129 mg (80% of theory) of the protected intermediate were obtained as a colourless foam.
Subsequently, the 129 mg (380 µmol) were deblocked with 2 ml of trifluoroacetic acid in 8 ml of dichloromethane. After stirring at RT for 1 h, the reaction mixture was concentrated under reduced pressure. The residue was lyophilized from acetonitrile/water, which left 125 mg (83% of ) of the title compound as a colourless foam.
LC-MS (Method 1): Rt = 0.38 min; MS (ESIpos): m/z = 240 (M+H)+ Intermediate 23 N-(2-aminoethyl)(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-N-methylbutanamide trifluoroacetate CH O H x H N N First, 35 mg (164 µmol) of tert-butyl 2-(methylamino)ethyl carbamate hydrochloride trifluoroacetate, 30 mg (164 µmol) of 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoic acid, 75 mg (197 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 57 µl of isopropylethylamine were ed in 5 ml of DMF and stirred at RT ght. Subsequently, the solvent was removed under reduced pressure and the remaining residue was purified by means of preparative HPLC. The corresponding fractions were concentrated and, by lyophilization from dioxane/water, 35 mg (63% of ) of the protected intermediate were obtained.
HPLC (Method 12): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.71 min; MS (ESIpos): m/z = 340 (M+H)+.
Subsequently, the entire amount of the protected ediate was deblocked with 1 ml of trifluoroacetic acid in 5 ml of dichloromethane to obtain 28 mg (77% of theory) of the title compound.
LC-MS (Method 3): Rt = 0.75 min; MS (ESIpos): m/z = 240 (M+H)+. 2012/075277 Intermediate 24 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-N-[2-(methylamino)ethyl]butanamide trifluoroacetate H C N 3 N N CF3COOH x H First, 35 mg (164 µmol) of tert-butyl noethyl)methyl carbamate hydrochloride trifluoroacetate, 30 mg (164 µmol) of 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoic acid, 75 mg (197 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 57 µl of N,N-diisopropylethylamine were combined in 5 ml of DMF and stirred at RT for 30 min. Subsequently, the solvent was removed under reduced pressure and the remaining residue was purified by means of preparative HPLC. The corresponding fractions were trated and, by lyophilization from dioxane/water, 51 mg (91% of theory) of the protected intermediate were obtained.
HPLC (Method 12): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.77 min; MS (ESIpos): m/z = 340 (M+H)+.
Subsequently, the entire amount was deprotected with 1 ml of trifluoroacetic acid in 5 ml of dichloromethane to obtain 45 mg (69% of theory) of the title compound.
LC-MS d 1): Rt = 0.19 min; MS s): m/z = 240 (M+H)+.
Intermediate 25 benzyl (2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoate trifluoroacetate CF3COOH x O O O First, (2R,3R)[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoic acid was released from 1.82 g (388 mmol) of its dicyclohexylamine salt by taking it up in ethyl acetate and extractive shaking with 100 ml of 0.5% sulphuric acid. The organic phase was dried over magnesium sulphate, filtered and concentrated. The residue was taken up in 10 ml of dioxane and WO 87716 ml of water, 1517 mg (4.66 mmol) of caesium carbonate were added, and the mixture was treated in an ultrasound bath for 5 min and concentrated under reduced pressure and redistilled once with DMF. The residue was then taken up in 15 ml of dichloromethane, and 1990 mg (11.64 mmol) of benzyl bromide were added. The mixture was treated in an ultrasound bath for 15 min and then concentrated under reduced pressure. The residue was partitioned between ethyl e and water, and the c phase was removed and extracted by shaking with saturated sodium chloride solution and then concentrated. The residue was then purified by preparative HPLC. This gave 1170 mg (80% of theory) of the Boc-protected intermediate.
Subsequently, these 1170 mg were deprotected immediately with 5 ml of trifluoroacetic acid in 15 ml of dichloromethane. After stirring at RT for 15 min, the reaction e was concentrated under reduced pressure. The residue was lyophilized from dioxane. After drying under high vacuum, there remained 1333 mg (84% of theory) of the title compound as a yellow oil.
HPLC (Method 6): Rt = 1.5 min; LC-MS (Method 1): Rt = 0.59 min; MS (ESIpos): m/z = 278 (M+H)+.
Intermediate 26 N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 CH O O 3 H OH H C N N 3 O N N 3 O O CH O CH O O CH 3 3 CH 3 H C CH 3 3 3 1200 mg (2.33 mmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(2R,3S,4S)carboxy methoxymethylhexanyl]-N-methyl-L-valinamide (Intermediate 5) were combined with 910.8 mg (2.33 mmol) of benzyl (2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoate trifluoroacetate (Intermediate 14), 1327 mg (3.49 mmol) of O-(7-azabenzotriazolyl)-N,N,N',N'- tetramethyluronium hexafluorophosphate and 2027 µl of N,N-diisopropylethylamine in 50 ml of DMF, and the mixture was d at RT for 5 min. Thereafter, the solvent was removed under reduced pressure. The remaining residue was taken up in ethyl acetate and extracted by shaking successively with 5% aqueous citric acid solution and saturated sodium hydrogencarbonate on. The organic phase was removed and concentrated. The residue was ed by means of preparative HPLC. The product ons were ed and concentrated, and the residue was dried under high vacuum. This gave 1000 mg (55% of theory) of the benzyl ester intermediate N- (tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)(benzyloxy) methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide as a resin.
LC-MS (Method 1): Rt = 1.56 min; MS (ESIpos): m/z = 775 (M+H)+.
The entire amount of this intermediate obtained was taken up in 25 ml of a mixture of methanol and dichloromethane (20:1), and the benzyl ester group was d by hydrogenation under standard en re with 10% palladium on activated carbon as a catalyst. After stirring at RT for 30 min, the catalyst was filtered off and the filtrate was concentrated under reduced pressure. This gave 803 mg (91% of theory) of the title compound as a white solid.
HPLC (Method 6): Rt = 2.1 min; LC-MS (Method 1): Rt = 1.24 min; MS (ESIpos): m/z = 685 (M+H)+.
Intermediate 27 )aminophenyl-N-propylcyclopropanecarboxamide trifluoroacetate HN 3 2 O CF3COOH x The title compound was prepared by ng commercially available (1S,2R)[(tertbutoxycarbonyl ]phenylcyclopropanecarboxylic acid with n-propylamine in the presence of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and subsequent Boc detachment with trifluoroacetic acid (yield: 85% of theory over both stages).
HPLC (Method 6): Rt = 1.2 min; LC-MS (Method 1): Rt = 0.52 min; MS (ESIpos): m/z = 219 (M+H)+.
Intermediate 28 ethyl (1S,2R)aminophenylcyclopropanecarboxylate trifluoroacetate O CH CF3COOH x O The title compound was prepared by standard methods, by esterifying commercially available (1S,2R)[(tert-butoxycarbonyl)amino]phenylcyclopropanecarboxylic acid with l and subsequent Boc detachment with trifluoroacetic acid.
LC-MS (Method 1): Rt = 0.50 min; MS s): m/z = 206 (M+H)+.
Intermediate 29 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-2,2-dimethylbutanoic acid H C CH 3 3 O To a solution of 1.39 g (8.95 mmol) of N-methoxycarbonylmaleimide in 44 ml of ted sodium hydrogencarbonate solution were added, at 0°C, 1.5 g (8.95 mmol) of 4-amino-2,2-dimethylbutyric acid, and the mixture was stirred for 40 min. Subsequently, the g bath was removed and the reaction mixture was stirred for a further 1 h. While cooling with ice, the reaction mixture was then adjusted to pH 3 by adding sulphuric acid, and ted with ethyl acetate. The combined organic phases were dried over magnesium sulphate and concentrated. 1.17 g (79% purity, 49% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.64 min; m/z = 212 (M+H)+.
Intermediate 30 tert-butyl 2-[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-2,2-dimethylbutanoyl]hydrazinecarboxylate H C CH O 3 3 O H C H 3 O N N HH C CH O 3 3 O To a solution of 50 mg (237 µmol) of 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-2,2- dimethylbutanoic acid in 2 ml of THF were added, at 0°C, first 26 µl (237 µmol) of 4- methylmorpholine and then 31 µl (237 µmol) of isobutyl chloroformate. After removing the cooling bath and stirring at RT for a further 15 min, 31.3 mg (237 µmol) of tert-butyloxycarbonyl hydrazide were added. The on e was stirred overnight and then concentrated. The residue was ed by preparative HPLC. 50.8 mg (66% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.71 min; m/z = 324 (M-H)-.
Intermediate 31 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-2,2-dimethylbutanehydrazide trifluoroacetate CF3COOH x H N N 2 N HH C CH 3 3 O 50 mg (154 mmol) of utyl 2-[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-2,2- dimethylbutanoyl]hydrazinecarboxylate were dissolved in 2 ml of dichloromethane, and 0.4 ml of trifluoroacetic acid was added. The reaction mixture was d at RT for 30 min and then concentrated. 55.2 mg (93% purity, 99% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.36 min; m/z = 226 (M+H)+.
Intermediate 32 adamantanylmethyl N-(tert-butoxycarbonyl)-L-phenylalaninate CH HN H C O 3 O O To a on of 500 mg (1.89 mmol) of N-Boc-L-phenylalanine in 25 ml of dichloromethane were added, at RT, 1192 mg (6.2 mmol) of EDC, 578 µl (4.1 mmol) of triethylamine, 345 mg (2.8 mmol) of DMAP and 345 mg (2.1 mmol) of 1-adamantylmethanol. The reaction mixture was stirred overnight, then diluted with 50 ml of dichloromethane, and was successively washed with % aqueous citric acid on, water and saturated sodium chloride solution. The c phase was dried over magnesium sulphate, then concentrated, and the residue was purified by preparative HPLC. 769 mg (90% of theory) of the title compound were obtained.
LC-MS (Method 2): Rt = 1.84 min; m/z = 414 (M+H)+.
Intermediate 33 adamantanylmethyl L-phenylalaninate hydrochloride HCl x H N 769 mg (1.86 mmol) of adamantanylmethyl N-(tert-butoxycarbonyl)-L-phenylalaninate (Intermediate 13) were dissolved in 25 ml of a 4 N solution of hydrogen chloride in dioxane and stirred at RT for 1 h. Subsequently, the reaction mixture was concentrated and the e was dried under reduced pressure. 619 mg (95% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.82 min; m/z = 314 .
Intermediate 34 N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) ntanylmethoxy)oxophenylpropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide WO 87716 2012/075277 H C CH H C CH 3 3 O 3 3 CH O O H 3 H N H C N N O 3 O N N 3 O O CH O CH3 O O CH 3 CH3 3 H C CH3 To a solution of 20 mg (29 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide in 1 ml of DMF were added, at RT, 15.3 µl (88 µmol) of N,N- diisopropylethylamine, 6.7 mg (44 µmol) of HOBt and 6.7 mg (35 µmol) of EDC, and the mixture was stirred for 30 min. Subsequently, 10.1 mg (32 µmol) of adamantanyl L-phenylalaninate hydrochloride were added. After stirring overnight, the reaction mixture was separated directly into its components via preparative HPLC. 27.5 mg (93% of theory) of the title compound were obtained.
LC-MS d 1): Rt = 1.70 min; m/z = 980 (M+H)+.
Intermediate 35 N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(adamantanylmethoxy)oxo phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C 3 CH 3 3 O 3 O H H N N N O HN N O O CH O CH O O CH 3 3 H C CH CH 3 3 3 3 27.5 mg (28 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(adamantanylmethoxy)oxophenylpropanyl]amino}methoxy methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide were dissolved in 1.8 ml of dichloromethane, and 361 µl of TFA were added. The reaction mixture was stirred for 30 min and then concentrated. The residue was taken up in water and lyophilized. 22.7 mg (81% of theory) of the title compound were obtained. 2012/075277 LC-MS d 1): Rt = 1.14 min; m/z = 880 (M+H)+. ediate 36 tert-butyl (2S)(benzyloxy)phenylpropanyl carbamate CH HN 3 O O Under an argon atmosphere, 500 mg (1.99 mmol) of N-Boc-L-phenylalaninol were dissolved in 5 ml of DMF and cooled to 0°C. Subsequently, 159 mg (3.98 mmol) of a 60% suspension of sodium hydride in paraffin oil were added. The reaction mixture was stirred until the evolution of gas had ended and then 260 µl (2.19 mmol) of benzyl bromide were added. The cooling bath was removed and the reaction mixture was stirred at RT for 2 h. Thereafter, the reaction mixture was concentrated, the e was taken up in ice-water and the mixture was extracted with dichloromethane. The organic phase was washed with saturated sodium chloride solution, dried over ium sulphate and concentrated. The residue was purified by means of preparative HPLC. 226 mg (33% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 1.28 min; m/z = 342 (M+H)+.
Intermediate 37 (2S)(benzyloxy)phenylpropanamine hydrochloride x HCl 220 mg (644 µmol) of tert-butyl (2S)(benzyloxy)phenylpropanyl carbamate were dissolved in 11 ml of a 4 N solution of hydrogen chloride in dioxane and d at RT for 1 h.
Then the reaction mixture was concentrated and the residue was dried under reduced pressure. 138 mg (77% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.65 min; m/z = 242 .
Intermediate 38 N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) (benzyloxy)phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH3 H C 3 CH 3 3 CH O O H 3 H N H C N N O 3 O N N 3 O O CH O CH3 O O CH 3 3 H C CH CH 3 3 3 To a solution of 20 mg (29 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide in 1 ml of DMF were added, at RT, 15.3 µl (88 µmol) of N,N- diisopropylethylamine, 6.7 mg (44 µmol) of HOBt and 6.7 mg (35 µmol) of EDC, and the mixture was stirred for 30 min. Subsequently, 7.8 mg (32 µmol) of (2S)(benzyloxy)phenylpropan amine hydrochloride were added. After stirring overnight, the reaction mixture was separated directly into its components via preparative HPLC. 26 mg (98% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 1.51 min; m/z = 909 (M+H)+.
Intermediate 39 N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)phenylpropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan methyl-L-valinamide oroacetate H C CH H C CH 3 3 3 3 O H H N N N O HN N O O CH O CH O O CH 3 3 H3C CH 3 3 3 x CF3COOH 26 mg (29 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)- 3-{[(2S)(benzyloxy)phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidin- 1-yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were ved in 1.8 ml of dichloromethane, and 370 µl of TFA were added. The reaction mixture was stirred at RT for 30 min and then concentrated. The residue was taken up in water and lyophilized. 26.4 mg (quant.) of the title compound were obtained.
LC-MS d 1): Rt = 0.97 min; m/z = 809 (M+H)+.
Intermediate 40 N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)hydroxyphenylpropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan methyl-L-valinamide H3C CH H C 3 3 CH3 OH O H H N N N HN N O O CH CH3 O CH O O 3 3 CH H C CH CH 3 3 3 3 50 mg (70 µmol) of Intermediate 26 and 11 mg (70 µmol) of (1S, 2R)aminophenylpropan ol in 10 ml of DMF were admixed with 42 mg (0.11 µmol) of O-(7-azabenzotriazolyl)- N,N,N',N'-tetramethyluronium hexafluorophosphate and 25 µl of N,N-diisopropylethylamine, and the reaction e was stirred at RT for 5 min. This was ed by concentration and purification of the residue by means of ative HPLC. After combining the corresponding fractions, concentrating and drying under high vacuum, 49 mg (81%) of the protected intermediate were obtained. Subsequently, the Boc group was detached by known conditions with trifluoroacetic acid in dichloromethane. Concentration was followed by the purification of the title compound by preparative HPLC, and 26 mg (52%) of the title compound were obtained.
HPLC (Method 12): Rt = 1.65 min; LC-MS (Method 1): Rt = 0.77 min; MS (ESIpos): m/z = 718 (M+H)+.
Intermediate 41 3-{2-[2-(2-aminoethoxy)ethoxy]ethoxy}propanoic acid trifluoroacetate O OH O 2 x CF3COOH 150 mg (541 µmol) of tert-butyl 3-{2-[2-(2-aminoethoxy)ethoxy]ethoxy}propanoate were dissolved in 3 ml of dichloromethane, 1.5 ml of trifluoroacetic acid were added, and the reaction mixture was stirred at RT for 1 h, then concentrated. 181 mg (100% of theory) of the title nd were obtained.
MS (EI): m/z 222 (M+H)+ ediate 42 3-(2-{2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethoxy)propanoic acid O OH O O 186 mg (555 µmol) of 3-{2-[2-(2-aminoethoxy)ethoxy]ethoxy}propanoic acid trifluoroacetate were dissolved in 2.6 ml of saturated sodium hydrogencarbonate solution and admixed at 0°C with 86 mg (555 µmol) of N-methoxycarbonylmaleimide. The reaction mixture was stirred at 0°C for 40 min and at RT for 1 h, then cooled again to 0°C, adjusted to pH 3 with sulphuric acid and extracted 3x with 25 ml of ethyl acetate. The combined organic phases were dried over magnesium sulphate and trated. 126 mg (75% of theory) of the title compound were ed.
LC-MS (Method 1): Rt = 0.53 min; m/z = 302 (M+H)+.
Intermediate 43 tert-butyl 5-dioxo-2,5-dihydro-1H-pyrrolyl)oxo-7,10,13-trioxa-2,3-diazapentadecan oate H C 3 O 3 CH3 N O O N O O O 125 mg (417 µmol) of 3-(2-{2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethoxy) propanoic acid were dissolved at 0°C in 2.1 ml of THF and admixed with 46 µl (417 mmol) of 4- methylmorpholine and 54.5 µl (417 µmol) of isobutyl chloroformate. The ice bath was removed and the reaction mixture was stirred at RT for 30 min. Subsequently, at 0°C, 55 mg (417 µmol) of tert-butyloxycarbonyl ide were added. The reaction e was warmed to RT overnight, concentrated and purified by preparative HPLC. 60 mg (33% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.66 min; m/z = 416 (M+H)+.
Intermediate 44 3-(2-{2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethoxy)propanehydrazide trifluoroacetate O N O O x CF3COOH 60 mg (145 µmol) of tert-butyl 15-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)oxo-7,10,13-trioxa- 2,3-diazapentadecanoate were dissolved in 1 ml of dichloromethane, and 0.2 ml of trifluoroacetic acid was added. The reaction mixture was d at RT for 30 min and then concentrated. 62 mg (100% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.35 min; m/z = 316 (M+H)+.
Intermediate 45 benzyl (1S,2R)aminophenylcyclopropanecarboxylate trifluoroacetate 2 O CF3COOH x The title compound was ed by standard methods, by esterifying commercially available (1S,2R)[(tert-butoxycarbonyl)amino]phenylcyclopropanecarboxylic acid with benzyl l and subsequent Boc detachment with trifluoroacetic acid.
LC-MS (Method 1): Rt = 0.72 min; MS s): m/z = 268 (M+H)+.
Intermediate 46 N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S)carboxy- 2-phenylethyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 O CH O O H 3 H N H C N N OH 3 O N N 3 O O CH O CH O O CH 3 3 3 H C CH CH 3 3 3 383 mg (0.743 mmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(2R,3S,4S)carboxy methoxymethylhexanyl]-N-methyl-L-valinamide (Intermediate 8) were combined with 485 mg (0.743 mmol) of benzyl N-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-L- phenylalaninate trifluoroacetate (Intermediate 12), 424 mg (1.114 mmol) of O-(7-azabenzotriazol- 1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 388 µl of N,N-diisopropylethylamine in 15 ml of DMF, and the mixture was stirred at RT for 10 min. Subsequently, the solvent was removed under reduced re. The remaining residue was taken up in ethyl acetate and extracted by shaking successively with 5% s citric acid on and saturated sodium hydrogencarbonate solution. The organic phase was removed and concentrated, and the residue was ed by means of preparative HPLC. The product fractions were combined and trated, and the residue was dried under high vacuum. 335 mg (48% of theory) of the benzyl ester intermediate were obtained as a foam.
LC-MS (Method 1): Rt = 1.49 min; MS (ESIpos): m/z = 922 (M+H)+. 100 mg (0.11 mmol) of this intermediate were taken up in 15 ml of methanol and the benzyl ester group was d by hydrogenation under standard hydrogen pressure with 10% palladium on activated carbon as a catalyst. After stirring at RT for 1 h, the catalyst was filtered off and the filtrate was concentrated under reduced pressure. After lyophilization from dioxane, 85 mg (94% of theory) of the title compound were obtained as a solid.
HPLC (Method 12): Rt = 2.4 min; LC-MS (Method 1): Rt = 1.24 min; MS (ESIpos): m/z = 832 (M+H)+.
Intermediate 47 N-benzyl-L-tryptophanamide oroacetate 2 O H 202 mg (0.5 mmol) of 2,5-dioxopyrrolidinyl t-butoxycarbonyl)-L-tryptophanate and 45 mg (0.42 mmol) of benzylamine were ved in 10 ml of DMF, and 110 µl (630 µmol) of N,N- diisopropylethylamine were added. The reaction mixture was stirred at RT for 3 h. Subsequently, the reaction mixture was concentrated under reduced pressure and the residue was purified by flash chromatography on silica gel (eluent: 20:0.5:0.05 dichloromethane/methanol/17% aq. ammonia). The corresponding fractions were combined and concentrated. The resulting residue was digested with diethyl ether and then dried under high vacuum. Subsequently, this residue was taken up in 10 ml of dichloromethane, and 3 ml of anhydrous trifluoroacetic acid were added.
After stirring at RT for 45 minutes, the mixture was concentrated and the residue was purified by preparative HPLC. After drying under high vacuum, 117 mg (57% of theory over both stages) of the title compound were obtained.
HPLC (Method 12): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.66 min; MS (ESIpos): m/z = 294 (M+H)+.
Intermediate 48 (1S,2R)aminophenylcyclopropanecarboxamide oroacetate 50 mg (180 µmol) of commercially available (1S,2R)[(tert-butoxycarbonyl)amino] phenylcyclopropanecarboxylic acid were dissolved in 5 ml of DMF, 94 µl (541 µmol) of N,N- diisopropylethylamine, 31 mg (270 µmol) of N-hydroxysuccinimide and 41.5 mg (216 µmol) of EDC were added, and then the mixture was stirred at RT overnight. The reaction mixture was then concentrated, the residue was taken up in dioxane, 71 mg (901 µmol) of ammonium hydrogencarbonate were added, and the reaction mixture was then left to stand at RT for 3 days.
The reaction mixture was then diluted with a 1:1 mixture of ethyl e and water. The organic phase was removed, dried over magnesium sulphate and concentrated. The resulting residue was subsequently taken up in 3 ml of dichloromethane, and 3 ml of ous trifluoroacetic acid were added. Stirring at RT for 1 h was followed by concentration. The e was stirred with pentane, ed off with suction and lyophilized from dioxane. In this way, 32 mg (62% of theory over both stages) of the title compound were obtained.
HPLC (Method 6): Rt = 0.38 min; LC-MS (Method 1): Rt = 0.20 min; MS (ESIpos): m/z = 177 (M+H)+.
Intermediate 49 Nα-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-L-tryptophanamide trifluoroacetate CH3 NH2 CF3COOH x N O O O The title compound was prepared in analogy to the synthesis of Intermediate 13 from Starting Compound 1 and tophanamide hydrochloride.
HPLC (Method 5): Rt = 1.4 min; LC-MS (Method 1): Rt = 0.92 min; MS (ESIpos): m/z = 473 (M+H)+.
Intermediate 50 4-nitrophenyl -dioxo-2,5-dihydro-1H-pyrrolyl)ethyl carbamate O O N N 813 mg (3.1 mmol) of triphenylphosphine were dissolved in 25 ml of THF and cooled to -70°C under argon. After dropwise addition of 627 mg (3.1 mmol) of diisopropyl azodicarboxylate, the e was stirred for 5 min. Subsequently, 500 mg (3.1 mmol) of tert-butyl 2-aminoethyl carbamate dissolved in 5 ml of THF were added dropwise, and the reaction mixture was stirred at - 70°C for a further 5 min. Then 136.6 mg (1.55 mmol) of 2,2-dimethylpropanol dissolved in 1 ml of THF and 301 mg (3.1 mmol) of maleimide were added, the reaction mixture was stirred at - 70°C for a further 10 min and then the e was warmed to RT. After ng at RT for a further 16 h, the solvent was removed under reduced pressure and the residue was purified by means of ative HPLC. This gave 463 mg (62%) of the protected intermediate.
After removing the Boc protecting group under standard conditions, 652 mg of 1-(2-aminoethyl)- 1H-pyrrole-2,5-dione were obtained as the trifluoroacetate. 112.9 mg (543 µmol) of nitrophenyl chloroformate were dissolved in 30 ml of THF and, after addition of 100 mg (271.6 µmol) of 1-(2-aminoethyl)-1H-pyrrole-2,5-dione trifluoroacetate, the mixture was d at RT for 30 min. The e was filtered and the filtrate was concentrated to dryness and then slurried with diethyl ether. After filtration with suction and drying, 60 mg (95% of theory) of the title compound were obtained.
HPLC d 5): Rt = 0.65 min; LC-MS (Method 1): Rt = 0.74 min; MS (ESIpos): m/z = 306 (M+H)+.
Intermediate 51 -phenyl(5-phenyl-1,3,4-oxadiazolyl)ethanamine trifluoroacetate CF3COOH x 200 mg (0.75 mmol) of N-(tert-butoxycarbonyl)-L-phenylalanine were initially charged at 0°C in .5 ml of dichloromethane, and 128 mg (0.79 mmol) of 1,1'-carbonyldiimidazole were added. After min, 103 mg (0.75 mmol) of l hydrazide were added. After a further 45 min at 0°C, 500 mg (1.5 mmol) of carbon romide and 395 mg (1.5 mmol) of triphenylphosphine were finally added. The reaction mixture was stirred first at 0°C for 2 h and then at RT overnight. The mixture was subsequently concentrated on a rotary ator, and the residue was dried under high . The crude product thus obtained was purified by means of preparative HPLC. 217 mg (78% of theory) of the Boc-protected intermediate tert-butyl (1S)phenyl(5-phenyl-1,3,4- oxadiazolyl)ethyl carbamate were obtained.
LC-MS (Method 12): Rt = 1.15 min; MS (ESIpos): m/z = 366 (M+H) + 217 mg (0.59 mmol) of this intermediate were taken up in 3 ml of dichloromethane, 0.6 ml of trifluoroacetic acid were added, and the e was stirred at RT for 30 min. Subsequently, the reaction mixture was concentrated under reduced pressure. The remaining residue was the reaction mixture dried further under reduced pressure and then lyophilized from dioxane. In this way, 214 mg (90% of theory) of the title compound were obtained.
LC-MS (Method 11): Rt = 0.62 min; MS (ESIpos): m/z = 266 (M+H) + 2012/075277 Intermediate 52 (1R)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethanamine trifluoroacetate CF3COOH x N N 200 mg (0.75 mmol) of t-butoxycarbonyl)-D-phenylalanine were lly charged at 0°C in 5.5 ml of dichloromethane, and 128.3 mg (0.79 mmol) of 1,1'-carbonyldiimidazole were added.
After 30 min, 103 mg (0.75 mmol) of benzoyl hydrazide were added. After a further 45 min at 0°C, 500 mg (1.5 mmol) of carbon tetrabromide and 395 mg (1.5 mmol) of triphenylphosphine were finally added. The reaction mixture was stirred first at 0°C for 2 h and then at RT ght. The mixture was subsequently concentrated on a rotary evaporator, and the residue was dried under high vacuum. The crude product thus obtained was purified by means of preparative HPLC. 219 mg (80% of theory) of the Boc-protected intermediate tert-butyl (1R)phenyl(5-phenyl-1,3,4- oxadiazolyl)ethyl carbamate were obtained.
LC-MS (Method 2): Rt = 1.36 min; MS (ESIpos): m/z = 366 (M+H) + 219 mg (0.6 mmol) of this intermediate were taken up in 3 ml of romethane, 0.6 ml of trifluoroacetic acid were added, and the mixture was stirred at RT for 30 min. Subsequently, the reaction mixture was concentrated under reduced pressure. The remaining residue was the reaction mixture dried further under reduced pressure and then lyophilized from dioxane. In this way, 196 mg (86% of theory) of the title compound were obtained as a solid.
HPLC (Method 10): Rt = 2.41 min Intermediate 53 -(benzylsulphonyl)phenylpropanamine NH2 O 200 mg (1.13 mmol) of -benzyl-1,3-oxazolidinone were initially charged in 3 ml of tertbutanol , and 280 mg (2.26 mmol) of benzyl mercaptan were added. The mixture was subsequently 2012/075277 heated under reflux for 2 days. Thereafter, the reaction mixture was concentrated on a rotary evaporator and the resulting (2S)(benzylsulphanyl)phenylpropanamine ediate was converted further directly, without .
HPLC (Method 10): Rt = 2.63 min LC-MS (Method 1): Rt = 0.67 min; MS (ESIpos): m/z = 258 (M+H) + The crude intermediate obtained above was dissolved in a solution of 2 ml of 30% hydrogen peroxide and 5 ml of formic acid, and the mixture was stirred at RT for 12 h. Then the reaction e was added to saturated sodium sulphate on and extracted three times with ethyl acetate. The organic phase was dried over magnesium sulphate and concentrated under reduced pressure. The crude product ed was purified by means of preparative HPLC. 343 mg (61% of theory) of the title compound were thus obtained.
HPLC (Method 10): Rt = 2.40 min; LC-MS (Method 12): Rt = 0.65 min; MS (ESIpos): m/z = 290 (M+H) + Intermediate 54 (2S,3E)-1,4-diphenylbutenamine 552.7 mg (9.85 mmol) of potassium hydroxide were dissolved in methanol, adsorbed onto 1.1 g of neutral aluminium oxide and then dried under high vacuum. To a solution of 240 mg (0.82 mmol) of (2S)(benzylsulphonyl)phenylpropanamine and 1.56 g of the potassium hydroxide on aluminium oxide thus prepared in 6.2 ml of n-butanol were added dropwise, at 5-10°C, 307 µl (3.3 mmol) of dibromodifluoromethane. The reaction mixture was stirred at RT for 2 h, then filtered through , and the residue was washed thoroughly with dichloromethane. The filtrate was concentrated and the resulting residue was dried under reduced re. The crude product thus obtained was purified by means of ative HPLC. 98 mg (35% of theory) of the title compound were obtained with an E/Z diastereomer ratio of 4:1.
HPLC (Method 10): Rt = 2.46 min; LC-MS (Method 12): Rt = 0.75 min; MS (ESIpos): m/z = 224 (M+H) + The E/Z diastereomer mixture obtained above was dissolved in 2 ml of ethanol and 0.2 ml of N,N- diisopropylethylamine, and separated by means of HPLC on chiral phase [column: Daicel Chiralpak AD-H, 5 µm 250 mm x 20 mm, eluent: hexane/(ethanol + 0.2% diethylamine) 50:50 v/v; UV detection: 220 nm; temperature: 30°C]. The appropriate fractions were concentrated on a rotary evaporator, and the residue was dried under reduced pressure. 45 mg of the title nd were obtained. 1H NMR (400 MHz, DMSO-d 6) δ [ppm] = 2.62 - 2.83 (m, 2 H) 3.52 - 3.71 (m, 1 H) 6.18 - 6.30 (m, 1 H) 6.34 - 6.46 (m, 1 H) 6.98 - 7.57 (m, 10 H) [further signals hidden under solvent peaks].
Intermediate 55 yl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo {[(1S)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H3C CH H C 3 3 CH3 O O H N H N x CF3COOH N N N HN N O O CH O CH 3 O O CH3 3 H C CH3 3 CH 20 mg (29 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)- 2-carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide were dissolved in 1 ml of DMF, 13.3 mg (35 µmol) of HATU and 15.3 µl (88 µmol) of N,N-diisopropylethylamine were added, and the mixture was stirred at RT for 30 min.
Subsequently, 12.2 mg (32 µmol) of (1S)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethanamine trifluoroacetate were added. The reaction e was stirred at RT overnight and then separated by preparative HPLC. This gave 22 mg (81% of ) of N-(tert-butoxycarbonyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl- 1-(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptan methyl-L-valinamide.
LC-MS (Method 12): Rt = 1.45 min; MS (ESIpos): m/z = 933 (M+H)+ By subsequently detaching the BOC protecting group with trifluoroacetic acid, 22.4 mg (98% of ) of the title compound were then obtained.
LC-MS (Method 11): Rt = 0.85 min; MS s): m/z = 833 (M+H)+ Intermediate 56 N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo {[(1R)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidinyl}methyl- 1-oxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH3 H C 3 CH O O 3 3 H N H N N N N x CF3COOH HN N O O CH O CH O O CH 3 3 3 H C CH 3 3 CH N-(tert-Butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) ymethyloxo{[(1R)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide was prepared in analogy to the synthesis of Intermediate 55, by reaction of 20 mg (29 µmol) of N-(tertbutoxycarbonyl )-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide with 12.2 mg (32 µmol) of (1R)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethanamine trifluoroacetate.
Yield: 17 mg (64% of theory) HPLC (Method 10): Rt = 3.74 min; LC-MS (Method 1): Rt = 1.45 min; MS (ESIpos): m/z = 933 (M+H) + By subsequently detaching the BOC protecting group with trifluoroacetic acid, 17.1 mg (99% of ) of the title compound were then obtained.
HPLC (Method 10): Rt = 2.55 min; LC-MS (Method 11): Rt = 0.85 min; MS (ESIpos): m/z = 833 (M+H) + Intermediate 57 N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzylsulphonyl)phenylpropan- 2-yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide trifluoroacetate 3 O S H C CH H C 3 3 3 CH O 3 O H N N N x CF3COOH HN N O O CH O CH O CH 3 H C CH 3 O 3 3 3 CH N-(tert-Butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) (benzylsulphonyl)phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide was prepared in analogy to the synthesis of Intermediate 55, by reaction of 20 mg (29 µmol) of N-(tert-butoxycarbonyl)-N-methyl- L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide with 9.3 mg (20 µmol) of (2S) lsulphonyl)phenylpropanamine.
Yield: 19.2 mg (68 % of theory) HPLC d 10): Rt = 3.5 min; LC-MS d 12): Rt = 1.41 min; MS (ESIpos): m/z = 957 (M+H) + By subsequently detaching the BOC protecting group with trifluoroacetic acid, 19.3 mg (99% of theory) of the title compound were then obtained.
HPLC (Method 10): Rt = 2.52 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 857 (M+H) + Intermediate 58 N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3E)-1,4-diphenylbuten yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide trifluoroacetate H C CH H C 3 3 3 CH O 3 H H N N N x H HN N O O CH O CH O O CH 3 H C CH 3 3 3 CH N-(tert-Butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3E)-1,4- diphenylbutenyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy oxoheptanyl]-N-methyl-L-valinamide was prepared in analogy to the synthesis of Intermediate 55, by reaction of 20 mg (29 µmol) N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N- [(3R,4S,5S){(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyl- 1-oxoheptanyl]-N-methyl-L-valinamide with 7.1 mg (32 µmol) of (2S,3E)-1,4-diphenylbuten- 2-amine.
Yield: 15.1 mg (58 % of theory) HPLC (Method 10): Rt = 4.2 min; LC-MS (Method 12): Rt = 1.51 min; MS (ESIpos): m/z = 891 (M+H) + By subsequently ing the BOC protecting group with trifluoroacetic acid, 15.7 mg (99% of theory) of the title compound were then obtained.
HPLC (Method 10): Rt = 2.62 min; LC-MS (Method 12): Rt = 0.97 min; MS (ESIpos): m/z = 791 (M+H) + Intermediate 61 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 N O 3 H H N HO N N O N N O O O CH O CH O O H C 3 3 3 H C CH CH 3 3 3 50 mg (0.054 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate mediate 16) were dissolved in 8 ml of dioxane/water, and 70 ml (0.108 mmol) of a 15% solution of 4-oxobutanoic acid in water were added. The reaction e was subsequently stirred at 100°C for 1 h. After cooling to RT, 3.7 mg (0.059 mmol) of sodium cyanoborohydride were added and the mixture was adjusted to a pH of 3 by adding about 300 µl of 0.1 N hydrochloric acid. The reaction mixture was then stirred at 100°C for a further 2 h. After cooling, another 70 ml (0.108 mmol) of the 15% utanoic acid solution were added and the reaction mixture was again stirred at 100°C for 1 h. Then a further 3.7 mg (0.059 mmol) of sodium cyanoborohydride were added and about 300 µl of 0.1 N hydrochloric acid were uently used to adjust the pH back to 3. The reaction mixture was then d at 100°C for another 2 h. In the event of conversion still being incomplete, this procedure was repeated for a third time. The on mixture was finally concentrated and the residue was purified by means of preparative HPLC. In this way, 32 mg (65% of theory) of the title compound were obtained in the form of a colourless foam.
HPLC (Method 5): Rt = 1.64 min; LC-MS (Method 9): Rt = 4.76 min; MS (ESIpos): m/z = 899 (M+H)+ 1H NMR (500 MHz, DMSO-d6): δ = 8.95 and 8.8 (2m, 1H), 8.88 and 8.65 (2s, 1H), 7.4-7.1 (m, 5H), 5.0, 4.78, 4.65 and 4.55 (4m, 2H), 4.1-3.7 (m, 5H), 3.32, 3.29, 3.20, 3.12, 3.1 and 3.0 (6s, 9H), 2.75 (m, 2H), 2.63 (t, 1H), 2.4-2.2 (m, 4H), 2.1-1.2 (m, 12H), 1.2-0.8 (m, 16H), 0.75 (m, 3H) er signals hidden under H2O and DMSO peaks].
Intermediate 62 arboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 O 3 N H N HO N N O N N O O O CH O CH O O H C 3 3 CH 3 H C CH 3 3 3 The title compound was prepared in analogy to the synthesis of Intermediate 61, by reaction of 50 mg of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl (1,2-oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidinyl} methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate (Intermediate 14) with 4- oxobutanoic acid.
Yield: 34 mg (70% of theory) HPLC (Method 5): Rt = 1.64 min; LC-MS (Method 9): Rt = 4.77 min; MS (ESIpos): m/z = 887 (M+H)+.
Intermediate 63 N-(4-carboxybenzyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 O 3 N H N N N O N N O O HO CH O CH O O H C 3 3 CH 3 H C CH 3 3 3 The title compound was prepared in analogy to the synthesis of Intermediate 61, by reaction of mg of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl {[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidin yl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate (Intermediate 16) with 4- formylbenzoic acid.
Yield: 7.5 mg (48% of theory) HPLC (Method 5): Rt = 1.75 min; LC-MS d 1): Rt = 0.97 min; MS s): m/z = 947 (M+H)+.
Intermediate 64 N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 N O H H N HO N N O N N O O O CH O CH O O H C 3 3 C CH CH 3 3 3 3 10 mg (0.011 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate (Intermediate 16) were ved in 2 ml of dioxane/water, and 2.8 mg (0.022 mmol) of 6- 2012/075277 oxohexanoic acid were added. The reaction mixture was subsequently stirred at 100°C for 1 h.
After cooling to RT, 0.75 mg (0.012 mmol) of sodium cyanoborohydride was added and the mixture was adjusted to a pH of 3 by adding 0.1 N hydrochloric acid. The reaction mixture was then stirred at 100°C for a further hour. After cooling, r 2.8 mg (0.022 mmol) of 6- oxohexanoic acid were added and the reaction e was again stirred at 100°C for 1 h. A further 0.75 mg (0.012 mmol) of sodium cyanoborohydride was added and 0.1 N hydrochloric acid was subsequently used to adjust the pH back to 3. The reaction mixture was then stirred at 100°C for another 1 h. This procedure was then repeated for a third time. The reaction mixture was finally concentrated and the crude product was purified by means of preparative HPLC. This gave 6.4 mg (64% of theory) of the title compound in the form of a less foam.
HPLC (Method 5): Rt = 1.68 min; LC-MS (Method 9): Rt = 4.86 min; MS s): m/z = 927 (M+H)+.
Intermediate 65 N-(2-aminoethyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide fluoroacetate H3C CH H C 3 CH 3 O 3 N H N H N N N 2 O N N O O CH O CH O O H C 3 3 3 H C CH3 CH 3 3 x 2 CF3COOH The title compound was prepared by reaction of 68 mg of N-methyl-L-valyl-N-[(3R,4S,5S) methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)(1,2-oxazinanyl)oxo phenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide trifluoroacetate (Intermediate 14) with tert-butyl 2-oxoethyl ate and subsequent detachment of the Boc protecting group with trifluoroacetic acid.
Yield: 49 mg (62% of theory over two stages) HPLC (Method 5): Rt = 1.58 min; LC-MS (Method 2): Rt = 1.05 min; MS (ESIpos): m/z = 844 (M+H)+ 1H NMR (600 MHz, DMSO-d6): δ = 8.25 (m, 1H), 8.45 and 8.15 (2d, 1H), 7.65-7.55 (m, 3H), 7.23-7.1 (m, 5H), 5.12 and 4.95 (2m, 1H), 4.72 and 4.62 (2m, 1H), 4.6 and 4.52 (2t, 1H), 4.2-3.8 (m, 4H), 3.7 (d, 1H), 3.23, 3.20, 3.19, 3.18, 3.03 and 2.98 (6s, 9H), 7 (m, 6H), 2.4-1.2 (m, 15H), 1.05, 1.0, 0.88 and 0.82 (4d, 6H), 0.92 (m, 6H), 0.73 (m, 6H) er signals hidden under H2O peak].
Intermediate 66 N-(3-aminopropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 N O 3 H N N N O H N N N 2 O O CH O CH O O H C 3 3 H C CH CH 3 3 3 3 The title compound was prepared in analogy to the synthesis of Intermediate 65, by reaction of mg (0.027 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate (Intermediate 16) with benzyl 3-oxopropyl carbamate and subsequent hydrogenolytic detachment of the Z protecting group (with 10% palladium on charcoal as a catalyst, in ethanol as a solvent).
Yield: 11 mg (41% of theory over two stages) HPLC (Method 5): Rt = 1.53 min; LC-MS d 1): Rt = 0.72 min; MS (ESIpos): m/z = 870 (M+H)+.
Intermediate 67 arboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(adamantan- 1-ylmethoxy)oxophenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 CH O 3 3 3 O H H N O N N O N N O O OH CH O CH O O CH 3 3 3 H C CH CH 3 3 3 26 mg (26 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(adamantan ylmethoxy)oxophenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate and 33.9 µl of a 15% aqueous aldehydic acid solution (53 µmol) were dissolved in 957 µl of a 1:1- dioxane/water mixture and heated to 100°C for 1 h. After brief cooling, 1.81 mg (29 µmol) of sodium cyanoborohydride were added. The reaction mixture was adjusted to pH 3 by adding 0.1 N hydrochloric acid and the mixture was heated to 100°C for a r 2 h. After again adding the same amounts of succinaldehydic acid solution, sodium cyanoborohydride and hydrochloric acid, the mixture was heated once again to 100°C for 2 h. The reaction mixture was then separated ly into its components by means of preparative HPLC. 18.5 mg (73% of theory) of the title compound were thus obtained.
LC-MS d 1): Rt = 1.17 min; m/z = 967 (M+H)+.
Intermediate 68 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)- ylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 O H H N O N N O N N O O OH CH O CH O O CH 3 3 3 H C CH CH 3 3 3 24 mg (26 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy) phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate and 33.7 µl of a 15% aqueous succinaldehydic acid solution (52 µmol) were dissolved in 953 µl of a 1:1-dioxane/water mixture and heated to 100°C for 1 h. After brief cooling, 1.80 mg (29 µmol) of sodium cyanoborohydride were added. The reaction mixture was adjusted to pH 3 by adding 0.1 N hydrochloric acid and the e was heated to 100°C for a further 2 h. After again adding the same amounts of succinaldehydic acid solution, sodium cyanoborohydride and hloric acid, the mixture was heated once again to 100°C for 2 h. The reaction mixture was then separated directly into its components by means of preparative HPLC. 15.2 mg (65% of ) of the title compound were thus obtained.
LC-MS (Method 1): Rt = 1.01 min; m/z = 895 (M+H)+ Intermediate 69 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)- 1-oxophenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy- 5-methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH O 3 3 3 3 O H H N O N N O N N O O OH CH O CH O O CH 3 3 H C CH CH 3 3 3 3 53 mg (84 µmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S) carboxymethoxymethylhexanyl]-N-methyl-L-valinamide mediate 4) and 45 mg (84 µmol) of benzyl N-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-L- alaninate trifluoroacetate (Intermediate 12) were taken up in 2 ml of DMF, 19 µl of N,N- diisopropylethylamine, 14 mg (92 µmol) of HOBt and 17.6 mg (92 µmol) of EDC were added and then the mixture was stirred at RT overnight. Subsequently, the reaction mixture was concentrated and the residue was purified by means of preparative HPLC. This gave 59 mg (68% of ) of the Fmoc-protected intermediate N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N- [(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)oxophenylpropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide.
LC-MS (Method 1): Rt = 1.55 min; m/z = 1044 (M+H)+. 57 mg (0.055 mmol) of this intermediate were treated with 1.2 ml of piperidine in 5 ml of DMF to detach the Fmoc protecting group. After concentration and purification by means of preparative HPLC, 39 mg (76% of theory) of the free amine intermediate N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R){[(2S)(benzyloxy)oxophenylpropanyl]amino}methoxymethyl- ropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were obtained as the trifluoroacetate.
HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 1.01 min; m/z = 822 . 37 mg (0.045 mmol) of this intermediate were dissolved in 5 ml of dioxane/water and, analogously to the preparation of the nd in Intermediate 66, d with a 15% aqueous on of 4- oxobutanoic acid in the presence of sodium cyanoborohydride. 16 mg (39% of theory) of the title compound were obtained in the form of a colourless foam.
HPLC (Method 6): Rt = 2.1 min; LC-MS (Method 1): Rt = 1.01 min; MS (ESIpos): m/z = 908 (M+H)+.
Intermediate 70 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3S) (benzyloxy)oxophenylbutanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}- 3-methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH 3 O O H H N O N N O N N O O OH CH O CH O O CH 3 3 3 H C H C CH CH 3 3 3 3 First, in analogy to the synthesis described in Intermediate 14, proceeding from Intermediates 4 and 26, the amine nd N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3S) (benzyloxy)oxophenylbutanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}- 3-methoxymethyloxoheptanyl]-N-methyl-L-valinamide was prepared. 30 mg (0.032 mmol) of this compound were dissolved in 6 ml of dioxane/water, and 41 µl (0.063 mmol) of a 15% aqueous solution of 4-oxobutanoic acid were added. The reaction mixture was subsequently stirred at 100°C for 1 h. After cooling to RT, 2.2 mg (0.035 mmol) of sodium cyanoborohydride were added and the mixture was adjusted to a pH of 3 by adding about 300 µl of 0.1 N hydrochloric acid. The reaction mixture was then d at 100°C for a further 2 h. After cooling, another 41 µl (0.063 mmol) of the 15% 4-oxobutanoic acid solution were added and the reaction mixture was again stirred at 100°C for 1 h. Then a further 2.2 mg (0.035 mmol) of sodium cyanoborohydride were added and about 300 µl of 0.1 N hloric acid were subsequently used to adjust the pH back to 3. The reaction mixture was then stirred at 100°C for another 2 h. In the event of conversion still being incomplete, this procedure was repeated for a third time. The reaction mixture was finally concentrated and the crude product was purified by means of preparative HPLC. This gave 24 mg (82% of theory) of the title compound in the form of a colourless foam.
HPLC (Method 5): Rt = 1.9 min; LC-MS d 9): Rt = 5.15 min; MS (ESIpos): m/z = 922 (M+H)+.
Intermediate 71 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy {[(2S)methoxyoxophenylpropanyl]amino}methyloxopropyl]pyrrolidinyl} methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 O O H H N CH N O 3 O N N N O O OH CH O CH O O CH 3 3 3 H C CH CH 3 3 3 First, in analogy to the synthesis described in Intermediate 14, ding from Intermediates 4 and 39, the amine compound N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxy{[(2S)methoxyoxophenylpropanyl]amino}methyl oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide was prepared. 7 mg (0.009 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the presence of sodium cyanoborohydride, to obtain 2 mg (22% of theory) of the title compound in the form of a colourless foam.
HPLC (Method 6): Rt = 1.9 min; LC-MS (Method 2): Rt = 1.06 min; MS (ESIpos): m/z = 832 (M+H)+.
Intermediate 72 arboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)- 3-(1H-indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide WO 87716 H C CH H C 3 3 3 CH 3 O O H H N O N N O N N O O OH CH O CH O O CH3 3 3 H C CH CH 3 3 3 212 mg (411 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(2R,3S,4S)carboxy methoxymethylhexanyl]-N-methyl-L-valinamide (Intermediate 8) and 237 mg (411 µmol) of benzyl-N-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-L-tryptophanate oroacetate (Intermediate 20) were taken up in 30 ml of DMF, and 188 mg (493 µmol) of O- (7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 215 µl N,N- diisopropylethylamine were added. The reaction mixture was stirred at RT for 20 h, then trated under reduced pressure, and the residue was purified by means of preparative HPLC.
The product fractions were ed and concentrated, and the e was dried under high vacuum. This gave 315 mg (80% of theory) of the Boc-protected intermediate N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy) (1H-indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide as a colourless foam.
LC-MS (Method 1): Rt = 1.45 min; m/z = 961 (M+H)+. 50 mg (52 µmol) of this intermediate were treated with 1 ml of trifluoroacetic acid in 9 ml of dichloromethane to detach the Boc protecting group. After concentration and purification by means of preparative HPLC, 29 mg (57% of theory) of the free amine intermediate N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)(1H-indolyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan methyl-L-valinamide were obtained as the trifluoroacetate.
LC-MS (Method 1): Rt = 0.99 min; m/z = 861 (M+H)+. 29 mg (0.03 mmol) of this intermediate were dissolved in 6 ml of dioxane/water, and 39 µl (0.059 mmol) of a 15% aqueous solution of 4-oxobutanoic acid were added. The reaction e was subsequently stirred at 100°C for 1 h. After cooling to RT, 2 mg (0.033 mmol) of sodium cyanoborohydride were added and the mixture was adjusted to a pH of 3 by adding about 300 µl of 0.1 N hydrochloric acid. The reaction mixture was then d at 100°C for a further 2 h. After cooling, another 39 µl (0.059 mmol) of the 15% 4-oxobutanoic acid solution were added and the reaction mixture was again stirred at 100°C for 1 h. Then a further 2 mg (0.033 mmol) of sodium 2012/075277 cyanoborohydride were added and about 300 µl of 0.1 N hydrochloric acid were subsequently used to adjust the pH back to 3. The mixture was then stirred at 100°C for another 2 h. Thereafter, the reaction mixture was poured onto a 1:1 mixture of semisaturated aqueous ammonium chloride solution and ethyl acetate. The organic phase was removed, washed with saturated sodium chloride solution, dried over sodium te and concentrated. The e was freeze-dried from water/ acetonitrile. This gave 27 mg (94% of ) of the title compound in the form of a colourless foam.
HPLC (Method 5): Rt = 2.2 min; LC-MS (Method 9): Rt = 5.04 min; MS (ESIpos): m/z = 947 (M+H)+.
Intermediate 73 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)({(2S) [benzyl(methyl)amino]oxophenylpropanyl}amino)methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide 3 H C H C CH H C CH 3 N 3 3 3 3 O H H N O N N O N N O O OH CH O CH O O CH 3 3 CH 3 H C CH 3 3 3 First, in analogy to the synthesis described in ediate 14, proceeding from Intermediates 4 and 38, the amine compound N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)({(2S) [benzyl(methyl)amino]oxophenylpropanyl}amino)methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide was prepared. 25 mg (0.026 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with utanoic acid in the presence of sodium cyanoborohydride, to obtain 13 mg (54% of theory) of the title compound in the form of a colourless foam.
HPLC (Method 12): Rt = 2.2 min; LC-MS (Method 9): Rt = 5.01 min; MS (ESIpos): m/z = 921 (M+H)+.
Intermediate 74 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)({(1S,2R) [(benzyloxy)carbonyl]phenylcyclopropyl}amino)methoxymethyloxopropyl]pyrrolidin- 1-yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 O O H H N O N N O N N O O OH CH O CH O O 3 CH 3 H C CH CH 3 3 3 3 50 mg (73 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)- 2-carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide (Intermediate 26) and 28 mg (73 µmol) of benzyl (1S,2R)amino phenylcyclopropanecarboxylate oroacetate (Intermediate 45) were taken up in 5 ml of DMF, and 42 mg (110 µmol) of zabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 38 µl of N,N-diisopropylethylamine were added. The reaction mixture was stirred at RT for 5 h, then concentrated under reduced pressure, and the residue was purified by means of preparative HPLC. The product fractions were combined and concentrated. After lyophilization from dioxane/water, 35 mg (51% of theory) of the Boc-protected intermediate N- (tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)({(1S,2R) [(benzyloxy)carbonyl]phenylcyclopropyl}amino)methoxymethyloxopropyl]pyrrolidin- 1-yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were obtained as a less foam.
LC-MS d 1): Rt = 1.52 min; m/z = 934 (M+H)+. 35 mg of this intermediate were treated with 1 ml of trifluoroacetic acid in 5 ml of romethane to detach the Boc protecting group. After concentration and lyophilization from dioxane/water, 34 mg (97% of theory) of the free amine intermediate N-methyl-L-valyl-N- [(3R,4S,5S){(2S)[(1R,2R)({(1S,2R)[(benzyloxy)carbonyl] phenylcyclopropyl}amino)methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide were obtained as the trifluoroacetate.
LC-MS d 1): Rt = 0.91 min; m/z = 834 (M+H)+. 11 mg (0.011 mmol) of this intermediate were then used, in analogy to the preparation of Intermediate 66, by reaction with utanoic acid in the presence of sodium cyanoborohydride, to obtain 2.5 mg (24% of theory) of the title compound in the form of a colourless foam.
HPLC (Method 12): Rt = 2.2 min; LC-MS d 9): Rt = 5.1 min; MS (ESIpos): m/z = 920 (M+H)+.
Intermediate 75 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyloxo{[(1S,2R)phenyl(propylcarbamoyl)cyclopropyl]amino}propyl]pyrrolidin yl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 3 HN O H H N O N N O N N O O OH CH O CH O O 3 3 CH H C CH CH 3 3 3 3 First, in analogy to the synthesis described in Intermediate 74, by coupling of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and )aminophenyl-N-propylcyclopropanecarboxamide trifluoroacetate (Intermediate 27) in the presence of O-(7-azabenzotriazolyl)-N,N,N',N'- tetramethyluronium hexafluorophosphate and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine nd N-methyl-L-valyl-N-[(3R,4S,5S)methoxy- 1-{(2S)[(1R,2R)methoxymethyloxo{[(1S,2R)phenyl (propylcarbamoyl)cyclopropyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-NL-valinamide was prepared as the trifluoroacetate. 14 mg (0.016 mmol) of this compound were then used, in analogy to the preparation of ediate 61, by reaction with 4-oxobutanoic acid in the presence of sodium cyanoborohydride, to obtain 11.3 mg (83% of theory) of the title compound.
HPLC (Method 6): Rt = 1.9 min; LC-MS (Method 2): Rt = 1.27 min; MS (ESIpos): m/z = 871 (M+H)+.
Intermediate 76 arboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R) (ethoxycarbonyl)phenylcyclopropyl]amino}methoxymethyloxopropyl]pyrrolidinyl}- 3-methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 O CH O H 3 H N HO N N O N N O O O CH O CH O O H C 3 3 H C CH CH 3 3 3 3 First, by coupling of Intermediate 46 (N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide) with Intermediate 48 (ethyl (1 1-amino phenylcyclopropanecarboxylate trifluoroacetate) in the presence of O-(7-azabenzotriazolyl)- N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent Boc ment, the starting compound N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)(ethoxycarbonyl) phenylcyclopropyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate was ed. 70 mg (0.079 mmol) of this starting material were then used, by reaction with 4-oxobutanoic acid, in analogy to Intermediate 61, to obtain 46 mg (68% of theory) of the title compound.
HPLC (Method 6): Rt = 1.9 min; LC-MS (Method 2): Rt = 1.28 min; MS (ESIpos): m/z = 858 (M+H)+ Intermediate 77 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)aminooxo- 3-phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH O 3 3 3 3 O H H N O N N NH N N 2 O O OH CH3 O CH O O CH H CH CH 3 3 3 3 First, in analogy to the synthesis described in Intermediate 75, by coupling of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide mediate 26) and L-phenylalaninamide hydrochloride in the presence of O-(7-azabenzotriazol- N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine compound N-methyl-L-valyl-N- [(3R,4S,5S){(2S)[(1R,2R){[(2S)aminooxophenylpropanyl]amino}methoxy- 2-methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide was prepared as the trifluoroacetate. 47 mg (0.049 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the presence of sodium orohydride, to obtain 39 mg (96% of theory) of the title nd.
HPLC (Method 6): Rt = 1.7 min; LC-MS (Method 9): Rt = 4.44 min; MS (ESIpos): m/z = 817 (M+H)+ 1H NMR (500 MHz, DMSO-d 6): δ = 8.95 and 8.8 (2m, 1H), 8.25 and 8.0 (2d, 1H), 7.45, 7.35 and 7.0 (3s, broad, 2H), 7.3-7.1 (m, 5H), 4.8-4.4 (2m, 3H), 3.95 (m, 1H), 3.82 (m, 1H), 3.72 (d, 1H), 3.22, 3.18, 3.15, 3.05 and 3.00 (5s, 9H), 2.85-2.7 (m, 4H), 2.45-1.6 (m, 12H), 1.5-1.2 (m, 3H), 1.1- 0.7 (m, 21H) [further signals hidden under solvent .
Intermediate 78 N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH N 3 3 3 O 3 H N H2N N N O N N O O CH3 O CH O O H C H C CH CH 3 3 3 3 This compound was prepared in analogy to Intermediate 66 over 2 stages, proceeding from 20 mg (16 µmol) of the compound from Intermediate 14 and benzyl 6-oxohexyl carbamate, and the hydrogenation was performed in methanol as the t.
Yield: 7.6 mg (55% of theory over 2 stages) HPLC (Method 6): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.7 min; MS (ESIpos): m/z = 901 (M+H)+.
Intermediate 79 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) (benzylamino)oxophenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidin methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 HN O H H N O N N O N N O O OH CH O CH O O CH 3 3 CH 3 H C CH 3 3 3 36 mg (43 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)- 3-{[(1S)carboxyphenylethyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 46) and 4.6 mg (43 µmol) of benzylamine were taken up in 5 ml of DMF, 7.5 µl (88 µmol) of N,N- ropylethylamine, 10 mg (65 µmol) of HOBt and 10 mg (52 µmol) of EDC were added, and then the mixture was stirred at RT overnight. Subsequently, the reaction mixture was concentrated and the residue was purified by means of ative HPLC. 29 mg (73% of theory) of the Boc- protected intermediate N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(benzylamino)oxophenylpropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were obtained.
LC-MS (Method 1): Rt = 1.43 min; m/z = 921 (M+H)+. 29 mg of this intermediate were treated with 1 ml of trifluoroacetic acid in 6 ml of dichloromethane to detach the Boc protecting group. After concentration and lyophilization from dioxane/water, 30 mg (quant.) of the free amine ediate N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R){[(2S)(benzylamino)oxophenylpropanyl]amino}methoxy methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide were obtained as the trifluoroacetate.
LC-MS (Method 1): Rt = 0.95 min; m/z = 821 (M+H)+. 17 mg (0.018 mmol) of this intermediate were then used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the presence of sodium orohydride, to obtain 13 mg (80% of theory) of the title nd in the form of a colourless foam.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 9): Rt = 4.97 min; MS (ESIpos): m/z = 907 (M+H)+.
Intermediate 80 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) (benzylamino)(1H-indolyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 CH HN 3 3 3 O H H N O N N O N N O O OH CH O CH O O CH3 3 3 H C CH CH 3 3 3 H First, in analogy to the synthesis described in Intermediate 74, by coupling of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and N-benzyl-L-tryptophanamide oroacetate (Intermediate 47) in the presence of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 2012/075277 uent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine compound N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzylamino)(1H- 3-yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} ymethyloxoheptanyl]-N-methyl-L-valinamide was prepared as the trifluoroacetate. 10 mg (0.01 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with utanoic acid in the presence of sodium cyanoborohydride, to obtain 2.5 mg (26% of theory) of the title compound.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 2): Rt = 1.13 min; MS (ESIpos): m/z = 946 (M+H)+. ediate 81 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R) carbamoylphenylcyclopropyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 CH3 3 3 NH O H 2 H N O N N O N N O O OH CH O CH O O 3 3 CH H C CH CH3 3 3 3 First, in analogy to the synthesis described in Intermediate 74, by coupling of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and (1S,2R)aminophenylcyclopropanecarboxamide trifluoroacetate (Intermediate 48) in the presence of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine compound N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(1S,2R)carbamoylphenylcyclopropyl]amino}methoxymethyloxopropyl]pyrrolidin- 1-yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide was prepared as the trifluoroacetate. 14 mg (0.0163 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the presence of sodium cyanoborohydride, to obtain 8 mg (57% of theory) of the title compound.
HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 9): Rt = 4.64 min; MS (ESIpos): m/z = 829 (M+H)+.
Intermediate 82 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H- 3-yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 NH O H 2 H N O N N O N N O O OH CH3 O CH3 O O CH H C CH CH 3 3 3 3 First, in analogy to the synthesis described in Intermediate 69, by coupling of N-[(9H-fluoren oxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)carboxymethoxymethylhexanyl]- N-methyl-L-valinamide (Intermediate 4) and Nα-{(2R,3R)methoxymethyl[(2S)-pyrrolidin- ropanoyl}-L-tryptophanamide trifluoroacetate (Intermediate 49) in the presence of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent ment of the Fmoc protecting group by means of piperidine, the amine compound N-methyl- L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropan no}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide was prepared as the trifluoroacetate. 78 mg (0.088 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4- oxobutanoic acid in the presence of sodium cyanoborohydride, to obtain 68 mg (90% of theory) of the title compound.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 9): Rt = 4.49 min; MS (ESIpos): m/z = 856 (M+H)+.
Intermediate 83 N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H- indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH O 3 NH H 2 H N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 OH H This compound was prepared in analogy to the compound in ediate 82, proceeding from 20 mg (26 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H- indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate, by reaction with 4- oxobutanoic acid in the presence of sodium cyanoborohydride prepared.
Yield: 5 mg (25% of theory) HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 11): Rt = 0.72 min; MS s): m/z = 884 (M+H)+.
Intermediate 84 arboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(2S)(morpholinyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidin yl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH N 3 3 3 O 3 H N HO N N O N N O O O CH O CH O O H C 3 3 C CH CH 3 3 3 3 First, in analogy to the synthesis described in Intermediate 79, by ng of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S)carboxy phenylethyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide (Intermediate 46) and morpholine in the presence of EDC and HOBT and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine compound N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyl{[(2S)(morpholinyl)oxophenylpropanyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide was prepared as the trifluoroacetate. 30 mg (0.033 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the ce of sodium cyanoborohydride, to obtain 22 mg (76% of theory) of the title compound.
HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 9): Rt = 4.58 min; MS (ESIpos): m/z = 887 (M+H)+.
Intermediate 85 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3R) (benzylamino)hydroxyoxobutanyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 CH 3 3 HN 3 H H N HO N N O N N O O O CH O CH O O H C 3 3 H C H C CH CH 3 3 OH 3 3 3 First, in analogy to the sis described in Intermediate 79, by coupling of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S)carboxy phenylethyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide (Intermediate 46) and N-benzyl-L-threoninamide trifluoroacetate in the presence of HATU and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine compound N-methyl-L-valyl-N-[(3R,4S,5S){(2S) R){[(2S,3R)(benzylamino)hydroxyoxobutanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide was prepared as the oroacetate. 21 mg (0.024 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the presence of sodium orohydride, to obtain 20 mg (97% of theory) of the title compound.
HPLC (Method 5): Rt = 1.54 min; LC-MS (Method 9): Rt = 4.49 min; MS (ESIpos): m/z = 861 (M+H)+.
Intermediate 86 4-{[(2S){[(2S){[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert.-Butoxyoxo propanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl](methyl)amino}methylbutanyl]amino}methyloxobutan yl](methyl)amino}butanoic acid CH 3 3 H C H C CH H C CH O CH 3 3 3 3 3 H N HO N N O N N O O O CH O CH O O H C 3 3 CH 3 H C CH 3 3 3 First, in y to the synthesis described in Intermediate 74, by coupling of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and tert-butyl-L-phenylalaninate hydrochloride in the presence of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid to obtain the tert-butyl ester (stirring with trifluoroacetic acid in dichloromethane for 40 minutes), the amine compound N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert-butoxyoxophenylpropan- 2-yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan- N-methyl-L-valinamide was prepared as the oroacetate. 22 mg (0.02 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4- oxobutanoic acid in the presence of sodium cyanoborohydride, to obtain 16 mg (94% of theory) of the title compound.
HPLC (Method 5): Rt = 2.0 min; LC-MS d 9): Rt = 5.05 min; MS (ESIpos): m/z = 874 (M+H)+.
Intermediate 87 4-{[(2S){[(2S){[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert-Butoxy(1H-indolyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl](methyl)amino}methylbutanyl]amino}methyloxobutan yl](methyl)amino}butanoic acid CH 3 3 H C H C CH H C CH 3 3 3 3 O CH H N HO N N O N N O O O CH O CH O O H C 3 3 3 H C CH CH 3 3 3 This compound was prepared in analogy to the synthesis described in Intermediate 86, proceeding from 230 mg (336 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N- methyl-L-valinamide (Intermediate 26) and utyl-L-tryptophanate hydrochloride over 3 stages.
Yield: 95 mg (31% of theory over 3 ) HPLC (Method 5): Rt = 2.0 min; LC-MS (Method 9): Rt = 5.05 min; MS (ESIpos): m/z = 913 (M+H)+.
Intermediate 88 N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H- indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH NH O 3 H 2 H N N N O N N O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 2 H First, in analogy to the syntheses described in Intermediate 69, by ng of N-[(9H-fluoren ylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)carboxymethoxymethylhexanyl]- N-methyl-L-valinamide (Intermediate 4) and Nα-{(2R,3R)methoxymethyl[(2S)-pyrrolidin- 2-yl]propanoyl}-L-tryptophanamide trifluoroacetate (Intermediate 49) in the ce of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent detachment of the Fmoc protecting group by means of piperidine, the amine compound N-methyl- L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide was prepared as the trifluoroacetate. 30 mg (0.03 mmol) of this compound were then used, in analogy to the preparation of the compound of Intermediate 61, by reaction with benzyl 6-oxohexyl carbamate, which had been obtained hand by oxidation of benzyl 6-hydroxyhexyl carbamate, in the presence of sodium cyanoborohydride, to obtain 17 mg (45% of ) of the Z-protected compound. Subsequently, hydrogenolysis in methanol over % palladium/activated carbon afforded the title compound.
Yield: 14 mg (95% of theory) HPLC (Method 5): Rt = 1.5 min; LC-MS (Method 1): Rt = 0.73 min; MS (ESIpos): m/z = 869 (M+H)+.
Intermediate 89 N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert-butoxy (1H-indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide CH 3 3 H C H C CH H C 3 3 3 CH 3 O CH O H 3 H N N N O N N O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 2 H First, in analogy to the synthesis described in Intermediate 86, by coupling of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and utyl-L-tryptophanate hydrochloride in the presence of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent ment of the Boc protecting group by means of trifluoroacetic acid to obtain the tert-butyl ester (stirring with 1:10 trifluoroacetic acid/dichloromethane for 30 min), the amine compound N- -L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert-butoxy(1H-indolyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide was prepared as the trifluoroacetate. 71 mg (0.075 mmol) of this nd were then used, in analogy to the preparation of the compound of ediate 61, by reaction with benzyl 6-oxohexyl carbamate, which had been obtained beforehand by ion of benzyl 6-hydroxyhexyl carbamate, in the presence of sodium cyanoborohydride, to obtain 35 mg (44% of theory) of the Z-protected compound. Subsequently, hydrogenolysis in methanol over 10% palladium/activated carbon afforded the title compound.
Yield: 30 mg (98% of theory) HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.77 min; MS (ESIpos): m/z = 926 (M+H)+.
Intermediate 90 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[2-(1H-indol yl)ethyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide H C CH3 H3C CH 3 O 3 H H N O N N N N O O OH CH O CH O O CH 3 3 H C CH CH 3 NH 3 3 3 First, in analogy to the synthesis described in Intermediate 74, by coupling of N-(tert- carbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and 2-(1H-indolyl)ethanamine in the presence of O-(7-azabenzotriazolyl)- N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine compound N-methyl-L-valyl-N- [(3R,4S,5S){(2S)[(1R,2R){[2-(1H-indolyl)ethyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide was prepared as the trifluoroacetate. 100 mg (0.119 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the presence of sodium cyanoborohydride, to obtain 50 mg (49% of theory) of the title compound. The title compound was purified here by flash tography on silica gel with dichloromethane/methanol/17% ammonia as the , in the course of which the mixing ratio was switched from initially 2 to 15/4/0.5.
HPLC (Method 6): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 813 (M+H)+.
Intermediate 91 N-(3-carboxypropyl)-N-methyl-L-valyl-N-{(3R,4S,5S)methoxy[(2S){(1R,2R)methoxy- 2-methyloxo[(2-phenylethyl)amino]propyl}pyrrolidinyl]methyloxoheptanyl}-N- methyl-L-valinamide H C CH H C CH 3 3 3 O 3 H H N O N N N N O O OH CH O CH O O 3 3 CH H C CH CH 3 3 3 3 First, in analogy to the synthesis described in Intermediate 74, by ng of N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and phenylethylamine in the ce of O-(7-azabenzotriazolyl)-N,N,N',N'- tetramethyluronium hexafluorophosphate and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine compound yl-L-valyl-N-{(3R,4S,5S) methoxy[(2S){(1R,2R)methoxymethyloxo[(2- phenylethyl)amino]propyl}pyrrolidinyl]methyloxoheptanyl}-N-methyl-L-valinamide was prepared as the trifluoroacetate. 57 mg (0.071 mmol) of this compound were then used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the ce of sodium cyanoborohydride, to obtain 44 mg (80% of theory) of the title compound. The title compound can also be purified here by flash tography on silica gel with dichloromethane/methanol/17% ammonia as the eluent (15/2/02 -> 15/4/0.5).
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 9): Rt = 4.64 min; MS (ESIpos): m/z = 774 (M+H)+.
Intermediate 92 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)hydroxy- 1-phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide 2012/075277 H C CH H C 3 3 3 CH 3 OH O H H N HO N N N N O O CH O CH O CH O O CH 3 3 3 H C CH CH 3 3 3 3 100 mg (0.139 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R) hydroxyphenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 40) were used, in analogy to the preparation of Intermediate 61, by reaction with 4-oxobutanoic acid in the presence of sodium cyanoborohydride, to obtain 94 mg (84% of theory) of the title compound. The title compound was purified by preparative HPLC.
HPLC (Method 5): Rt = 1.5 min; LC-MS (Method 9): Rt = 4.46 min; MS s): m/z = 804 (M+H)+.
Intermediate 93 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C O 3 3 O 3 CH 3 H N H N HO N N N N N O O O CH O CH3 O O CH 3 H C CH3 3 CH3 22.4 mg (24 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy- yloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate were dissolved in 1.4 ml of dioxane/water and, analogously to the ation of Intermediate 61, reacted with 15% aqueous solution of 4-oxobutanoic acid in the presence of sodium cyanoborohydride. After lyophilization from dioxane, 8.2 mg (38% of theory) of the title compound were obtained in the form of a white solid.
HPLC (Method 10): Rt = 2.54 min LC-MS (Method 12): Rt = 0.94 min; MS (ESIpos): m/z = 919 (M+H)+ Intermediate 94 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyloxo{[(1R)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide H3C CH3 H3C CH O O 3 H N H N HO N N N N N O O O CH O CH3 O O CH3 3 H C CH3 3 CH3 17.1 mg (18 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy- 2-methyloxo{[(1R)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate were ved in 1.1 ml of dioxane/water and, analogously to the preparation of Intermediate 61, reacted with 15% aqueous solution of utanoic acid in the ce of sodium cyanoborohydride. After lyophilization from dioxane, 14.8 mg (89% of theory) of the title compound were obtained in the form of a white solid.
HPLC (Method 10): Rt = 2.54 min; LC-MS (Method 12): Rt = 0.92 min; MS (ESIpos): m/z = 919 (M+H) + ediate 95 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) (benzylsulphonyl)phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide 3 O S H3C CH H3C 3 CH3 O O H N HO N N N N O O O CH O CH O CH 3 H C CH 3 O 3 3 3 CH 19.3 mg (20 µmol) N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) (benzylsulphonyl)phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate were ved in 1.2 ml of dioxane/water and, analogously to the preparation of Intermediate 61, reacted with % aqueous on of 4-oxobutanoic acid in the presence of sodium cyanoborohydride. After lyophilization from dioxane, 8.6 mg (45% of theory) of the title compound were obtained in the form of a solid.
LC-MS (Method 11): Rt = 0.85 min; MS (ESIpos): m/z = 943 (M+H) + Intermediate 96 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3E)-1,4- diphenylbutenyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide H C CH3 H C 3 3 CH O 3 H H N HO N N N N O O O CH O CH O CH 3 3 O 3 H C CH 3 3 CH 15.5 mg (10 µmol) of yl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3E)-1,4- diphenylbutenyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate were dissolved in 1.0 ml of 2012/075277 dioxane/water and, analogously to the preparation of Intermediate 61, reacted with 15% aqueous solution of 4-oxobutanoic acid in the presence of sodium orohydride. After lization from dioxane, 10.3 mg (68% of theory) of the title compound were obtained in the form of a white solid.
HPLC (Method 10): Rt = 2.59 min; LC-MS (Method 11): Rt = 0.94 min; MS s): m/z = 877 (M+H) + Intermediate 97 minohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 N O 3 H H N H N N N 2 O N N O O CH O CH O O H C 3 3 H C CH 3 3 3 3 The title compound was prepared in analogy to the synthesis of Intermediate 66, by reaction of 200 mg (0.108 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate (Intermediate 16) with benzyl 3-oxohexyl carbamate and subsequent hydrogenolytic detachment of the Z protecting group (with 5% palladium on charcoal as a catalyst, in methanol as a solvent).
Yield: 69 mg (65% of theory over two stages) HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.76 min; MS (ESIpos): m/z = 912 (M+H)+.
Intermediate 98 N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) (benzylamino)(1H-indolyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 CH 3 3 HN O H H N HO N N O N N O O O CH O CH O O H C 3 3 H C CH CH 3 NH 3 3 3 This compound was prepared in analogy to the synthesis described in Intermediate 80. The purification was ed by preparative HPLC.
Yield: 40 mg (29% of theory over 3 stages) HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.92 min; MS (ESIpos): m/z = 974 (M+H)+.
Intermediate 99 (2S)amino(1H-indolyl)(1,2-oxazinanyl)propanone trifluoroacetate CF3COOH x H N 2 O 324 mg (0.81 mmol) of 2,5-dioxopyrrolidinyl N-(tert-butoxycarbonyl)-L-tryptophanate were dissolved in 20 ml of DMF, and 200 mg (1.62 mmol) of 1,2-oxazinane hloride (Starting Compound 5) and 850 µl of N,N-diisopropylethylamine were added. The reaction mixture was d at 50°C overnight and then concentrated under d pressure. The residue was taken up in dichloromethane and extracted with water. The organic phase was dried over magnesium sulphate and concentrated. The residue was purified by flash tography on silica gel with 4:1 dichloromethane/ethyl acetate as the eluent. The product fractions were concentrated and the residue was dried under high vacuum. This gave 147.5 mg (48% of theory) of the Boc-protected intermediate.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 1.03 min; MS (ESIpos): m/z = 374 (M+H)+.
Using 166 mg (444.5 µmol) of this intermediate, under standard conditions with 3 ml of trifluoroacetic acid in 20 ml of dichloromethane, the Boc protecting group was detached and, after HPLC purification, 155 mg (86% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 1.43 min; LC-MS (Method 11): Rt = 0.56 min; MS (ESIpos): m/z = 274 (M+H)+.
Intermediate 100 [(benzyloxy)carbonyl]amino}hexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 CH 3 O 3 N H H N O N N N O N N O O O CH O CH O O H C 3 3 3 H C CH CH 3 3 3 177 mg (260 µmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-NL-valinamide (Intermediate 26) and 100 mg (260 µmol) of (2S)amino(1H-indolyl)- 1-(1,2-oxazinanyl)propanone trifluoroacetate (Intermediate 99) were taken up in 15 ml of DMF, and 118 mg (310 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 140 µl of N,N-diisopropylethylamine were added. The reaction mixture was stirred at RT for 30 min, then concentrated under d pressure, and the residue was purified by means of preparative HPLC. The product fractions were combined and concentrated.
After lyophilization from dioxane, 170 mg (68% of theory) of the Boc-protected ediate were obtained.
LC-MS (Method 1): Rt = 1.36 min; m/z = 940 (M+H)+. 170 mg of this intermediate were treated with 3 ml of oroacetic acid in 30 ml of dichloromethane for 30 min to detach the Boc protecting group. Then the reaction mixture was concentrated under reduced pressure and the residue was purified by means of preparative HPLC to obtain 155 mg (86% of ) of the deprotected N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxy oxopropyl] pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide intermediate.
HPLC (Method 12): Rt = 1.85 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 840 (M+H)+. 50 mg (0.052 mmol) of this intermediate were then used, in analogy to the preparation of Intermediate 97, with benzyl 3-oxohexyl carbamate in the presence of sodium cyanoborohydride and subsequent hydrogenolytic detachment of the Z ting group (with 5% palladium on charcoal as a catalyst, in methanol as a solvent), prepared to prepare the title nd.
Yield: 21 mg (37% of theory) HPLC d 12): Rt = 2.1 min; LC-MS (Method 1): Rt = 1.02 min; MS (ESIpos): m/z = 1073 (M+H)+.
Intermediate 101 N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)oxazinanyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH O 3 N H N H N N N 2 O N N O O CH O CH O O H C 3 3 H C CH 3 3 3 3 26.7 mg (24.87 µmol) of Intermediate 100 were dissolved in 10 ml of methanol and hydrogenated over palladium/activated carbon (5%) under standard hydrogen pressure for 30 min. The catalyst was filtered off and the solvent was evaporated off under d pressure. After the residue had been dried under high vacuum, 22.5 mg (96% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.76 min; MS (ESIpos): m/z = 939 (M+H)+.
Intermediate 102 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)(morpholin yl)oxophenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]- N-methyl-L-valinamide H C CH H C 3 3 O 3 O 3 N N H H N N N N O N N N O H O O O CH O CH O O 3 CH H C CH 3 CH 3 3 3 3 This compound was prepared in y to the synthesis described in Intermediate 157 from N-(3- carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy {[(2S)(morpholinyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidin yl}methyloxoheptanyl]-N-methyl-L-valinamide and commercially available 6-(2,5-dioxo- 2,5-dihydro-1H-pyrrolyl)hexanehydrazide.
Yield: 8 mg (71% of theory) HPLC d 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 1094 (M+H)+.
Intermediate 103 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3R)(benzylamino)hydroxyoxobutan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH HN O O 3 H N H H N N N N O N N N O H O O O CH O CH O O 3 3 CH OH 3 H C CH 3 H C CH 3 3 3 This compound was prepared in analogy to the synthesis bed in Intermediate 157 from N-(3- carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3R)(benzylamino)- 3-hydroxyoxobutanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy- -methyloxoheptanyl]-N-methyl-L-valinamide and commercially available 6-(2,5-dioxo-2,5- dihydro-1H-pyrrolyl)hexanehydrazide.
Yield: 3 mg (22% of theory) HPLC (Method 5): Rt = 1.6 min; LC-MS d 1): Rt = 0.78 min; MS (ESIpos): m/z = 1069 (M+H)+.
Intermediate 104 N-{4-[(trans{[(2,5-dioxopyrrolidinyl)oxy]carbonyl}cyclohexyl)amino]oxobutyl}-N- methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH3 NH O H 2 H H N N N N O O N N O O O O CH O CH O O N 3 3 CH H3C CH CH 3 3 3 O H First, benzyl transaminocyclohexanecarboxylate trifluoroacetate was prepared from trans aminocyclohexanecarboxylic acid by introducing the Boc protecting group, then introducing the benzyl ester protecting group and subsequently detaching the Boc protecting group by conventional peptide chemistry methods. mg (18 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyloxopropyl] pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were then ved in 5 ml of dimethylformamide and subsequently admixed with 13 mg (35 µmol) of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium uorophosphate, 9 µl of N,N- diisopropylethylamine and with 15 mg (44 µmol) of benzyl transaminocyclohexanecarboxylate trifluoroacetate. The e was d at RT for 1 h and then concentrated under reduced pressure. The remaining residue was purified by means of preparative HPLC. The corresponding fractions were combined and the solvent was evaporated off under reduced pressure. After the e had been dried under high vacuum, 14.7 mg (78% of theory) of the protected intermediate were obtained as a colourless foam.
HPLC (Method 6): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.95 min; MS (ESIpos): m/z = 1072 (M+H)+.
From this protected intermediate, the benzyl ester was first removed by hydrogenolytic means and the free carboxyl component was obtained in quantitative yield. 14 mg (14 µmol; 1 equiv.) of the deprotected compound were taken up in 5 ml of DMF and admixed with 3.3 mg (29 µmol; 2.1 equiv.) of N-hydroxysuccinimide in the presence of 4.1 mg (21 µmol; 1.5 equiv.) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 7.5 µl (44 µmol; 3.1 equiv.) of N,N- diisopropylethylamine and 0.9 mg (7 µmol; 0.5 equiv.) of thylaminopyridine, and the mixture was stirred at RT overnight. Then another 10 equiv. of N-hydroxysuccinimide, 5 equiv. of 1-(3-dimethylaminopropyl)ethylcarbodiimide hydrochloride, 5 equiv. of N,N- diisopropylethylamine and 0.5 equiv. of thylaminopyridine were added and the reaction mixture was treated in an ultrasound bath for 5 h. Subsequently, the solvent was evaporated off, the residue was purified by means of ative HPLC and the corresponding fractions were combined and concentrated. After lyophilization of the e from dioxane, 9.7 mg (62% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 6): Rt = 1.8 min; LC-MS (Method 11): Rt = 0.77 min; MS s): m/z = 1078 .
Intermediate 105 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S)carboxyphenylethyl]amino}methoxy methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide H C CH H C 3 3 3 CH 3 OH O O H N H H N N N N O N N N O H O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 This compound was prepared in analogy to the synthesis bed in Intermediate 157, proceeding from 4-{[(2S){[(2S){[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert-butoxyoxo phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl](methyl)amino}methylbutanyl]amino}methyloxobutan yl](methyl)amino}butanoic acid and commercially available -dioxo-2,5-dihydro-1H-pyrrol- 1-yl)hexanehydrazide. The ester intermediate was obtained in 42% yield. In a second step, 6 mg (6 µmol) of this intermediate were cleaved with trifluoroacetic acid the tert-butyl ester. After HPLC cation, 3.4 mg (48% of theory) of the title compound were obtained.
HPLC d 5): Rt = 1.66 min; LC-MS (Method 2): Rt = 1.04 min; MS (ESIpos): m/z = 1025 (M+H)+.
Intermediate 106 N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH3 NH O H 2 H N N N N O N N O O O CH3 O CH3 O O CH H3C CH3 CH 3 14 mg (16 µmol) of N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyloxopropyl] idinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 87) were taken up in 750 µl of dioxane and admixed with 1.5 ml of saturated sodium hydrogencarbonate solution and then with 3.2 mg (21 µmol) of methyl 2,5-dioxo-2,5-dihydro-1H- pyrrolecarboxylate. The reaction mixture was stirred at RT for 1 h and then concentrated under reduced pressure. The remaining e was purified by means of preparative HPLC. After lization, 5.5 mg (36% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.84 min; MS (ESIpos): m/z = 949 (M+H)+.
Intermediate 107 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[2-(1H-indolyl)ethyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH O O 3 H H O N N N N N N N H O HN O O CH O CH O O 3 H C H C CH CH 3 3 3 3 38 mg (47 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[2-(1H-indolyl)ethyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide were ved in 37 ml of DMF and then admixed with 71 mg (187 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 33 µl of N,N-diisopropylethylamine and with 37 mg (140 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was d at RT for 1 h. This was followed by tration under high vacuum and purification of the remaining residue by means of preparative HPLC. Thus, 12.2 mg (26% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.85 min; MS (ESIpos): m/z = 1020 (M+H)+.
Intermediate 108 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-{(3R,4S,5S)methoxy[(2S){(1R,2R)methoxymethyloxo[(2-phenylethyl) amino]propyl}pyrrolidinyl]methyloxoheptanyl}-N-methyl-L-valinamide O H C CH H C 3 3 3 CH O O 3 H H O N N N N N N N H O HN O O CH O CH O O H C 3 3 H C CH CH 3 3 3 3 The compound was prepared in analogy to Intermediate 107.
Yield: 2.5 mg (30% of theory) HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.9 min; MS (ESIpos): m/z = 981 .
Intermediate 109 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)hydroxyphenylpropanyl]amino} ymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide O H C CH H C 3 3 3 CH OH O O 3 H H N N N N N N N N H O O CH O O CH O CH O O CH 3 3 3 H C CH CH 3 3 3 3 The compound was prepared in analogy to Intermediate 107 from the compound in Intermediate 92.
Yield: 35 mg (65% of theory) HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 11): Rt = 0.76 min; MS (ESIpos): m/z = 1011 .
Intermediate 110 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide O O H C CH H C N 3 3 3 CH NH O 3 H 2 H N O N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 This compound was prepared in analogy to Intermediate 147 from the compound in Intermediate 83.
Yield: 2.4 mg (24% of theory) HPLC (Method 6): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 981 (M+H)+.
Intermediate 111 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]methylhydrazino}oxobutyl)-N- methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 CH O CH 3 O 3 3 H O N N N N O N N N H O HN O O CH O CH O O 3 H NH 3 C CH CH 3 2 3 3 3 This nd was prepared in analogy to Intermediate 140 from Intermediate 82 and Intermediate 22.
Yield: 6.5 mg (51% of theory) HPLC (Method 6): Rt = 1.8 min; LC-MS (Method 1): Rt = 4.71 min; MS (ESIpos): m/z = 1077 .
Intermediate 112 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)carbamoylphenylcyclopropyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide 2012/075277 O H C CH H C 3 3 3 CH NH O O 3 H 2 H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 This compound was ed in analogy to Intermediate 157 from the nd in Intermediate Yield: 5.7 mg (57 % of theory) HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 1036 (M+H)+.
Intermediate 113 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S)carboxy(1H-indolyl)ethyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide O H C CH H C 3 3 3 CH 3 OH O O H H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 95 mg (104 µmol) of 4-{[(2S){[(2S){[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert-butoxy- 3-(1H-indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl](methyl)amino}methylbutanyl]amino}methyl oxobutanyl](methyl)amino}butanoic acid were ved in DMF and then admixed with 79.5 mg (209 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 73 µl of N,N-diisopropylethylamine and with 68 mg (261 µmol) of commercially available 6-(2,5- dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was stirred at RT for 2 h. This was followed by concentration under high vacuum and purification of the remaining residue by means of preparative HPLC. Thus, 104 mg (89% of theory) of the tert-butyl ester of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.93 min; MS (ESIpos): m/z = 1121 (M+H)+.
The intermediate was taken up in 33.4 ml of dichloromethane, 17 ml of trifluoroacetic acid were added, and the mixture was stirred at RT for 1 h. Subsequently, the reaction mixture was concentrated under reduced re and the residue was purified by preparative HPLC.
Thus, 61 mg (62 % of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 1064 (M+H)+.
Intermediate 114 N-[6-({[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethyl]carbamoyl}amino)hexyl]-N-methyl-L-valyl- N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide H3C CH H3C O 3 CH3 NH O H 2 H H H N N N N N O N N N O O O CH O CH3 O O 3 H3C O H C CH CH3 3 3 mg (5 µmol) of minohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)- 1-amino(1H-indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidin- 1-yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were taken up in 885 µl of DMF and admixed with 5.3 mg (8 µmol) of 4-nitrophenyl 2-(2,5-dioxo-2,5-dihydro-1H-pyrrol yl)ethyl carbamate and 2.8 µl of N,N-diisopropylethylamine. The reaction mixture was d at RT for 2 h and then trated to dryness. The residue was purified by means of preparative HPLC.
Yield: 0.58 mg (11% of theory) of a colourless foam HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.83 min; MS (ESIpos): m/z = 1035 (M+H)+.
Intermediate 115 N-{4-[(2,5-dioxopyrrolidinyl)oxy]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl) phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide H C CH H C 3 3 3 CH N O O 3 H H N O N N O N N N O O O CH O CH O O 3 3 CH H C CH CH 3 O 3 3 3 This compound was prepared in analogy to the compound in Intermediate 147, proceeding from 8 mg (9 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl) phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- mide. After concentration, the activated ester was purified by means of preparative HPLC and, after removal of the solvent under reduced pressure, reacted immediately with the antibody.
Yield: 3 mg (27% of theory) (hydrolysis-sensitive) HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 996 (M+H)+.
Intermediate 116 (2,5-dioxopyrrolidinyl)oxy]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyl{[(2S)(1,2-oxazinanyl)oxophenylpropan yl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide WO 87716 H C CH H C 3 3 3 CH O 3 N O H H N O N N O N N N O O O CH O CH O O 3 3 CH3 O H3C CH CH 3 3 This compound was prepared in analogy to the compound in Intermediate 147, ding from 5 mg (6 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino}- 3-oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide. After concentration, the activated ester was purified by means of preparative HPLC and, after removal of the solvent under reduced pressure, reacted immediately with the antibody.
Yield: 3.2 mg (43% of theory) (hydrolysis-sensitive) HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.92 min; MS (ESIpos): m/z = 984 .
Intermediate 117 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert-butoxyoxophenylpropanyl]amino}- oxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N- methyl-L-valinamide CH3 3 CH O H C CH3 H C 3 3 CH3 O O O H H H N N N N N O N N N H O O O O CH3 O CH O O 3 H C H3C CH CH 3 3 3 This compound was prepared in analogy to Intermediate 157 from the compound in Intermediate 86.
Yield: 7 mg (42% of theory) WO 87716 HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.94 min; MS (ESIpos): m/z = 1081 (M+H)+.
Intermediate 118 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2R)(benzyloxy)phenylpropanyl]amino} ymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide O H C CH H3C 3 CH O 3 O 3 H H H N N N N N N N N H O O O O O CH O CH O O CH 3 H C CH3 3 3 CH3 The target compound was prepared analogously to ediate 157 from 7 mg (7.8 µmol) of the compound in Intermediate 68. Yield: 6.3 mg (53% of theory) LC-MS (Method 1): Rt = 1.00 min; MS (ESIpos): m/z = 1102 (M+H)+.
Intermediate 119 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl- 1-(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptan yl]-N-methyl-L-valinamide O H3C CH3 H3C CH3 O O O H N H H N N N N N N N N N H O O O O CH3 O CH3 O O CH3 H C CH3 3 CH 7.4 mg (8.1 mmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide and 6.3 mg (24.2 mmol) of 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide hydrochloride were coupled and worked up in analogy to Intermediate 157. 1.6 mg (13% of theory) of the title compound were obtained as a solid.
LC-MS (Method 11): Rt = 0.89 min; MS (ESIpos): m/z = 1126 (M+H) + Intermediate 120 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo{[(1R)phenyl- 1-(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptan yl]-N-methyl-L-valinamide O H3C CH H3C CH3 O O 3 O H N H H N N N N N N N N N H O CH3 O CH3O O O CH O O 3 H3C CH 3 CH3 12.8 mg (13.9 mmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)- 2-[(1R,2R)methoxymethyloxo{[(1R)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide and .9 mg (41.8 mmol) of 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide hydrochloride were d and worked up in analogy to Intermediate 157. 10.8 mg (59% of ) of the title compound were obtained as a solid.
LC-MS d 11): Rt = 0.90 min; MS (ESIpos): m/z = 1126 (M+H) + Intermediate 121 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzylsulphonyl)phenylpropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide 3 O O H C CH H C CH O 3 3 O 3 3 S H H N N N N N N N N H O O O O CH O CH O CH 3 O 3 3 H C CH 3 3 CH 7.4 mg (7.9 mmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) (benzylsulphonyl)phenylpropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide and 6.2 mg (23.5 mmol) of 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide hydrochloride were coupled and worked up in analogy to Intermediate 157. 6.9 mg (74% of theory) of the title compound were obtained as a solid.
LC-MS (Method 11): Rt = 0.87 min; MS s): m/z = 1150 (M+H) + Intermediate 122 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3E)-1,4-diphenylbutenyl]amino}methoxy- 2-methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide O H3C CH3 H3C CH3 O O H H H N N N N N N N N H O O O O CH O CH3 O O CH3 3 H C CH3 3 CH3 8 mg (9.1 mmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S,3E)-1,4-diphenylbutenyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide and 7.2 mg (27.4 mmol) of 6-(2,5- dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide hloride were coupled and worked up in analogy to Intermediate 157. 8.2 mg (82% of theory) of the title compound were obtained as a white solid.
LC-MS (Method 11): Rt = 0.95 min; MS (ESIpos): m/z = 1083 (M+H) + Intermediate 123 N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)tert-butoxy(1H-indolyl)oxopropanyl]amino}methoxymethyl- 3-oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide CH 3 3 H C O H C CH H C 3 3 3 CH O 3 O CH H 3 H N N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 H mg (30 µmol) of Intermediate 89 were taken up in 2 ml of 1,4-dioxane and admixed with 4 ml of saturated sodium hydrogencarbonate solution and then with 7.5 mg (50 µmol) of methyl 2,5- dioxo-2,5-dihydro-1H-pyrrolecarboxylate. The reaction mixture was stirred at RT for 1 h and then concentrated under reduced pressure. The remaining residue was ed by means of preparative HPLC. After lyophilization, 24 mg (74% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 2.2 min; LC-MS (Method 1): Rt = 1.01 min; MS (ESIpos): m/z = 1006 (M+H)+.
Intermediate 124 N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) R){[(1S)carboxy(1H-indolyl)ethyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH 3 OH O H H N N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 22 mg (20 µmol) of Intermediate 123 were d with 4 ml of trifluoroacetic acid in 8 ml of dichloromethane at RT for 1 h. Thereafter, the reaction e was concentrated under reduced re. The remaining residue was purified by means of preparative HPLC. After lyophilization, 11 mg (54% of theory) of the title compound were obtained.
WO 87716 HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 11): Rt = 0.85 min; MS (ESIpos): m/z = 950 (M+H)+.
Intermediate 125 2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxy methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide O H C CH H C 3 3 3 CH O 3 N H N N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 22.5 mg (20 µmol) of Intermediate 101 were taken up in 2 ml of 1:1 e/water and then admixed with 5.6 mg (40 µmol) of methyl 2,5-dioxo-2,5-dihydro-1H-pyrrolecarboxylate and with 0.25 ml of saturated sodium hydrogencarbonate solution. The reaction mixture was stirred at RT for 30 min. Then another 0.25 ml of the saturated sodium hydrogencarbonate solution was added and the reaction mixture was stirred at RT for a further 15 min and then trated under reduced re. The remaining residue was purified by means of preparative HPLC. After lyophilization, 12.8 mg (50% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.95 min; MS (ESIpos): m/z = 1019 (M+H)+.
Intermediate 126 N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl) phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide O H3C CH H C 3 3 CH3 N O H H N N N N O N N O O O CH O CH3 O O 3 CH3 H C CH CH 3 3 3 64 mg (70 µmol) of N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 97) were taken up in 3 ml of 1:1 dioxane/water, then adjusted to pH 9 with 4 ml of saturated sodium encarbonate solution and uently admixed with 16.3 mg (110 µmol) of methyl 2,5-dioxo-2,5-dihydro-1H-pyrrolecarboxylate. The reaction mixture was stirred at RT for 1 h and then concentrated under reduced pressure. Then another 8 mg (55 µmol) of methyl 2,5- dioxo-2,5-dihydro-1H-pyrrolecarboxylate were added, and the reaction mixture was adjusted again to pH 9 and stirred at RT for a further hour. This was followed by concentration and purification of the remaining residue by means of preparative HPLC. At first, 31 mg of an as yet uncyclized intermediate were obtained. 27 mg of this intermediate were taken up again in 2 ml of 1:1 dioxane/water and then admixed with 250 µl of saturated sodium encarbonate solution.
After stirring at RT for 2 hours, the on mixture was concentrated and the residue was purified by means of ative HPLC. After lyophilization, 20 mg (29% of theory) of the title compound were obtained.
HPLC d 5): Rt = 1.96 min; LC-MS (Method 1): Rt = 0.97 min; MS (ESIpos): m/z = 992 (M+H)+.
Intermediate 127 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(benzylamino)(1H-indolyl)oxopropanyl]amino}methoxy methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide H C CH H C O O 3 3 3 CH3 HN N O H H N O N N O N N O O O CH O CH O O 3 3 CH3 H C CH CH 3 3 3 17 mg (18 µmol) of N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)(benzylamino)(1H-indolyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 98) were ved in 2.8 ml of dichloromethane and admixed with 20 mg (174 mmol) of 1-hydroxypyrrolidine-2,5-dione and then with 10 mg (52 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride and 0.21 mg (0.17 µmol) of DMAP.
After stirring at RT for 4 h, the reaction mixture was concentrated under reduced pressure. The remaining residue was purified by means of preparative HPLC. After lyophilization, 8.2 mg (43% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.98 min; MS s): m/z = 1071 (M+H)+.
Intermediate 128 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)(1,2-oxazinan- 1-oxophenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptan yl]-N-methyl-L-valinamide O H C CH H C 3 3 CH 3 N O 3 O H H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 5 mg (5.6 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino}- 3-oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 845 µl of DMF and then d with 3.2 mg (17 µmol) of 1-(3-dimethylaminopropyl) ethylcarbodiimide hydrochloride, 2.6 mg (17 µmol) of 1-hydroxy-1H-benzotriazole hydrate, 1.96 µl of N,N-diisopropylethylamine and with 5.9 mg (22.5 µmol) of commercially available 6-(2,5- dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was stirred at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 2.2 mg (36% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.88 min; MS (ESIpos): m/z = 1094 (M+H)+.
Intermediate 129 N-(6-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxohexyl)-N-methyl-L- N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2- oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH N O O 3 H H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 4 mg (4.3 µmol) of N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 646 µl of DMF and then d with 2.5 mg (13 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 2.0 mg (13 µmol) of 1-hydroxy-1H- riazole hydrate, 2.25 µl of N,N-diisopropylethylamine and with 4.5 mg (17 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was stirred at RT for 3 h and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 1.9 mg (39% of theory) of the title nd were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 9): Rt = 4.9 min; MS (ESIpos): m/z = 1134 (M+H)+.
Intermediate 130 N-(4-{[(2R)({5-[(2,5-dioxopyrrolidinyl)oxy]oxopentanoyl}amino)propanyl]oxy} oxobutyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl 2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidin yl}methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C N O O 3 3 3 CH O 3 H N N O N N O O N N N H O O O CH O CH O CH O O CH 3 3 3 H C CH CH 3 3 3 3 .5 mg (11.7 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)- 2-[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 3.7 ml of dichloromethane and then d with 6.7 mg (35 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 0.7 mg (5.8 µmol) of 4- dimethylaminopyridine and with 8.2 mg (47 µmol) of commercially available tert-butyl (2R) hydroxypropyl ate. The mixture was stirred at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 7.5 mg (61% of theory) of the otected intermediate were obtained as a colourless foam.
HPLC (Method 5): Rt = 2.0 min; LC-MS d 1): Rt = 1.03 min; MS (ESIpos): m/z = 1056 (M+H)+.
Subsequently, the Boc protecting group was ed with trifluoroacetic acid. 4.9 mg (0.005 mmol) of the deprotected crude product were then, without further purification, taken up in 1.8 ml of dichloromethane and admixed with 3.7 mg (0.011 mmol) of 1,1'-[(1,5-dioxopentane-1,5- diyl)bis(oxy)]dipyrrolidine-2,5-dione, 2.4 µl (0.014 mmol) of N,N-diisopropylethylamine and 0.6 mg (5 µmol) of 4-dimethylaminopyridine. The mixture was d at RT for 2 h and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 0.77 mg (15% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.93 min; MS (ESIpos): m/z = 1167 (M+H)+.
Intermediate 131 N-{4-[(1-{5-[(2,5-dioxopyrrolidinyl)oxy]oxopentanoyl}piperidinyl)oxy]oxobutyl}-N- methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R) (1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl- 1-oxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH3 N O H H N O N N O O N N O O O N O CH O CH3 O O CH N 3 H C CH3 CH 3 3 3 O O mg (11 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 2 ml of dichloromethane and then admixed with 4.3 mg (22 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 0.88 mg (6 µmol) of 4- dimethylaminopyridine and with 5.2 mg (22 µmol) of commercially ble benzyl 4- hydroxypiperidinecarboxylate. The e was stirred at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 5 mg (40% of theory) of the Z-protected intermediate were obtained as a colourless foam.
HPLC (Method 5): Rt = 2.1 min; LC-MS d 1): Rt = 1.04 min; MS (ESIpos): m/z = 1116 (M+H)+.
Subsequently, the Z protecting group was detached by hydrogenolytic means in ethanol over palladium/activated carbon. 4.6 mg (0.005 mmol) of the deprotected crude product were then, without r purification, taken up in 1.8 ml of dichloromethane and admixed with 3.8 mg (0.012 mmol) of 1,1'-[(1,5-dioxopentane-1,5-diyl)bis(oxy)]dipyrrolidine-2,5-dione, 0.8 µl (0.005 mmol) of N,N-diisopropylethylamine and 0.6 mg (5 µmol) of 4-dimethylaminopyridine. The mixture was stirred at RT overnight and then trated under high vacuum. The remaining e was purified by means of preparative HPLC. Thus, 0.96 mg (16% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.94 min; MS (ESIpos): m/z = 1193 (M+H)+.
Intermediate 132 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazinyl}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2- anylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH N O O 3 H H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 mg (16.7 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 2500 µl of DMF and then admixed with 9.6 mg (50 µmol) of 1-(3- ylaminopropyl)ethylcarbodiimide hydrochloride, 7.6 mg (50 µmol) of 1-hydroxy-1H- benzotriazole hydrate, 5.8 µl of N,N-diisopropylethylamine and with 17.4 mg (67 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was stirred at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 11.2 mg (52% of theory) of the title compound were obtained as a less foam.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 2): Rt = 1.09 min; MS (ESIpos): m/z = 1106 (M+H)+.
Intermediate 133 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazinyl}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3S)(benzyloxy)oxophenylbutan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide O H C CH H C O 3 3 3 CH O O 3 H H H N N N N N O N N N H O O O O CH O CH O O 3 H C 3 H C CH CH 3 H C 3 3 3 3 .8 mg (6.3 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S,3S)(benzyloxy)oxophenylbutanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 943 µl of DMF and then admixed with 3.6 mg (19 µmol) of 1-(3- ylaminopropyl)ethylcarbodiimide hydrochloride, 2.9 mg (19 µmol) of 1-hydroxy-1H- benzotriazole hydrate, 2.2 µl of N,N-diisopropylethylamine and with 6.6 mg (25 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The e was stirred at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 4.5 mg (64% of theory) of the title compound were obtained as a less foam.
HPLC (Method 5): Rt = 2.0 min; LC-MS (Method 1): Rt = 1.03 min; MS (ESIpos): m/z = 1129 (M+H)+.
Intermediate 134 N-[3-({[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethyl]carbamoyl}amino)propyl]-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2- oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH3 N O O H H N N N N O N N N N H H O O O CH3 O CH3 O O H C H C CH CH3 3 3 3 First, 4-nitrophenyl -dioxo-2,5-dihydro-1H-pyrrolyl)ethyl carbamate was prepared under standard conditions, proceeding from commercially ble 1-(2-aminoethyl)-1H-pyrrole-2,5- dione trifluoroacetate and ophenyl carbonate. mg (6 µmol) of N-(3-aminopropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 1000 µl of DMF and then d with 2 µl of N,N-diisopropylethylamine and with 2.2 mg (9 µmol) of 4-nitrophenyl 2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethyl carbamate. The mixture was stirred at RT for 1 h and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 1.6 mg (23% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 2): Rt = 1.09 min; MS (ESIpos): m/z = 1036 (M+H)+.
Intermediate 135 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)oxophenylpropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide O H C CH H3C 3 3 CH3 O O O H H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH3 H C CH3 CH3 10 mg (11 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) (benzyloxy)oxophenylpropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 4000 µl of DMF and then admixed with 6.3 mg (33 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 4.5 mg (33 µmol) of 1-hydroxy-1H- benzotriazole hydrate, 5.7 µl of N,N-diisopropylethylamine and with 11.5 mg (44 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was stirred at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 2.6 mg (14% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 6): Rt = 2.1 min; LC-MS (Method 1): Rt = 1.01 min; MS (ESIpos): m/z = 1115 (M+H)+.
Intermediate 136 N-(4-{4-[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoyl]piperazinyl}oxobutyl)-N-methyl- L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2- oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide O CH N H C CH H C 3 CH N N 3 3 O 3 H N O N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 First, 1-[4-oxo(piperazinyl)butyl]-1H-pyrrole-2,5-dione trifluoroacetate was ed under standard conditions, proceeding from tert-butyl piperazinecarboxylate and 4-(2,5-dioxo-2,5- dihydro-1H-pyrrolyl)butanoic acid over 2 stages. mg (5.6 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 1000 µl of DMF and then admixed with 2.1 mg (11 µmol) of 1-(3- ylaminopropyl)ethylcarbodiimide hydrochloride, 1.7 mg (11 µmol) of 1-hydroxy-1H- benzotriazole hydrate, 2 µl of N,N-diisopropylethylamine and with 3.5 mg (5.6 µmol) of 1-[4-oxo- 4-(piperazinyl)butyl]-1H-pyrrole-2,5-dione trifluoroacetate. The mixture was stirred at RT ght. Then 2.1 mg (5.6 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate were added and the reaction mixture was d at RT for a further 3 h.
Subsequently, the solvent was removed under d pressure and the remaining residue was purified by means of preparative HPLC. The corresponding fractions were concentrated and, by lyophilization from water, 0.6 mg (10% of theory) of the title compound was obtained as a colourless foam.
HPLC (Method 6): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.9 min; MS (ESIpos): m/z = 1132 (M+H)+.
Intermediate 137 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]methylhydrazino}oxobutyl)-N- methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)(1,2- anyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH O 3 N CH O 3 H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 First, 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-N'-methylhexanehydrazide trifluoroacetate was prepared under rd conditions, proceeding from commercially available 6-(2,5-dioxo-2,5- dihydro-1H-pyrrolyl)hexanoic acid and tert-butyl ylhydrazinecarboxylate over 2 stages. 6.9 mg (8 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino}- 3-oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 2540 µl of DMF and then admixed with 3.6 mg (9 µmol) of O-(7-azabenzotriazolyl)- N,N,N',N'-tetramethyluronium hexafluorophosphate, 3 µl of N,N-diisopropylethylamine and with 4.1 mg (12 µmol) of 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-N'-methylhexanehydrazide trifluoroacetate. The e was stirred at RT overnight. Subsequently, the solvent was removed under reduced pressure and the remaining residue was purified by means of preparative HPLC.
Thus, 3.9 mg (45% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.93 min; MS (ESIpos): m/z = 1108 (M+H)+. ediate 138 N-{4-[(2-{[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoyl](methyl)amino}ethyl)(methyl) amino]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 CH N O CH3 3 O 3 H N N N N N O N N N O O O CH3 O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 Proceeding from tert-butylmethyl 2-(methylamino)ethyl carbamate and 4-(2,5-dioxo-2,5-dihydro- 1H-pyrrolyl)butanoic acid, over 2 stages, 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-N-methyl-N- [2-(methylamino)ethyl]butanamide trifluoroacetate was first prepared by. 6.6 mg (7.3 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl) phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide were dissolved in 2000 µl of DMF and then admixed with 5.6 mg (14.7 µmol) of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 2.6 µl of N,N- diisopropylethylamine and with 4.1 mg (9 µmol) of 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-N- methyl-N-[2-(methylamino)ethyl]butanamide trifluoroacetate. After stirring at RT for 3 h, the same amounts of HATU and N,N-diisopropylethylamine were added once again, and the reaction mixture was then d at RT overnight. Subsequently, the solvent was removed under reduced re and the remaining residue was purified by means of preparative HPLC. Thus, 4 mg (44% of ) of the title compound were obtained as a colourless foam.
HPLC (Method 6): Rt = 2.0 min; LC-MS d 1): Rt = 0.91 min; MS (ESIpos): m/z = 1134 .
Intermediate 139 (2R,3S)amino{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutan- 2-yl (3R,4S,7S,10S)[(2S)-butanyl]-7,10-diisopropyl(2-{(2S)[(1R,2R)methoxy methyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidin- 1-yl}oxoethyl)-5,11-dimethyl-6,9-dioxooxa-5,8,11-triazapentadecanoate O H C CH H C 3 CH N O NH 3 O 3 3 H H 2 H N N N O N N O N N N H O O O O CH O CH O CH O O 3 3 CH H C CH3 3 CH3 3 13 mg (14.7 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(2S)(1,2-oxazinanyl)oxophenylpropanyl]amino}- 3-oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 10 ml of dichloromethane and then d with 8.4 mg (44 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 5.4 mg (44 µmol) of 4- dimethylaminopyridine and with 9 mg (29.3 µmol) of commercially available benzyl N-(tert- butoxycarbonyl)-L-threoninate. The mixture was stirred at RT for 5 h. uently, the reaction mixture was twice ted by shaking with water and the c phase was dried over sodium sulphate and concentrated under reduced pressure. The ing residue was purified by means of preparative HPLC. After lyophilization from dioxane/water, 14 mg (81% of theory) of the protected intermediate were obtained as a colourless foam.
HPLC (Method 12): Rt = 2.3 min; LC-MS (Method 1): Rt = 1.13 min; MS (ESIpos): m/z = 1178 (M+H)+.
Subsequently, the Z protecting group was ed by hydrogenolytic means in methanol over % palladium/activated carbon. 9.5 mg (0.0087 mmol) of the deprotected crude product were then, without further purification, taken up in 5 ml of DMF, and admixed 5 mg (26.2 µmol) of 1- (3-dimethylaminopropyl)ethylcarbodiimide hydrochloride, 4 mg (26.2 µmol) of 1-hydroxy-1H- benzotriazole hydrate, 54.6 µl of N,N-diisopropylethylamine and with 9.1 mg (34.9 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was stirred at RT for 1 h and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. After lyophilization from dioxane, 9.5 mg (84% of theory) of the Boc-protected intermediate were obtained.
HPLC d 12): Rt = 2.1 min; LC-MS (Method 1): Rt = 0.97 min; MS (ESIpos): m/z = 1295 .
Subsequently, 9.5 mg (7.3 µmol) were deprotected with 0.5 ml of trifluoroacetic acid in 2 ml of dichloromethane of the Boc-protected intermediate and, after lyophilization from dioxane, 9 mg (82% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 12): Rt = 2.1 min; LC-MS (Method 1): Rt = 0.84 min; MS (ESIpos): m/z = 1195 (M+H)+.
Intermediate 140 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]methylhydrazino}oxobutyl)-N- methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R) (1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl- eptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH N O CH O 3 3 H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 4.1 mg (12 µmol) of 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-N'-methylhexanehydrazide trifluoroacetate (Intermediate 22) were dissolved together with 6.9 mg (8 µmol) of the compound from Intermediate 61 in 2.5 ml of DMF and then admixed with 3.5 mg (9 µmol) of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium uorophosphate and 3 µl of N,N- diisopropylethylamine. The mixture was d at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. After lyophilization from dioxane, 2.6 mg (30% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.90 and 0.91 min; MS (ESIpos): m/z = 1120 (M+H)+.
Intermediate 141 N-[4-({1-[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoyl]piperidinyl}oxy)oxobutyl]-N- methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R) (1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl- 1-oxoheptanyl]-N-methyl-L-valinamide 2012/075277 H C CH H C 3 3 3 CH O 3 N H N O N N O O N N O O N O CH O CH O O 3 3 CH N H C CH CH 3 3 3 3 44 mg (49 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 2 ml of dichloromethane and then admixed with 18.8 mg (98 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 3.8 mg (24 µmol) of 4- dimethylaminopyridine and with 23 mg (98 µmol) of commercially available benzyl 4- hydroxypiperidinecarboxylate. The mixture was stirred at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 22 mg (40% of theory) of the Z-protected intermediate were obtained as a colourless foam.
HPLC (Method 5): Rt = 2.1 min; LC-MS d 1): Rt = 1.04 min; MS (ESIpos): m/z = 1116 (M+H)+.
Subsequently, the Z protecting group was detached by hydrogenolytic means in l over palladium/activated carbon. 19 mg (19 µmol) of the ected crude product were then, t further purification, taken up in 4 ml of DMF and admixed with 7 mg (39 µmol) of 4-(2,5-dioxo-2,5-dihydro-1H-pyrrol yl)butanoic acid, 11 mg (29 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 5 µl of N,N-diisopropylethylamine. The e was stirred at RT for 1 h and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. After lyophilization from dioxane, 7.5 mg (34% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.94 min; MS (ESIpos): m/z = 1147 (M+H)+.
Intermediate 142 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)(1H-indolyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide O H C CH H C O 3 3 3 CH O O 3 H H H N N N N N O N N N H O O O O CH O CH O O 3 3 H C H C CH CH 3 3 3 3 9 mg (9.5 µmol) of arboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)(benzyloxy)(1H-indolyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 72) were dissolved in 1000 µl of DMF and then admixed with 10 mg (38 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide, 7.2 mg (19 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 8 µl of N,N-diisopropylethylamine, and the reaction mixture was stirred at RT for 1 h. Subsequently, the solvent was removed under reduced pressure and the remaining e was purified by means of preparative HPLC. The corresponding ons were concentrated and, by lyophilization, 6.4 mg (58% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.99 min; MS s): m/z = 1154 (M+H)+.
Intermediate 143 N-(4-{2-[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-2,2-dimethylbutanoyl]hydrazino}oxobutyl)- N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R)- 1-(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl} methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 CH N O O 3 3 H H C H H N N 3 N N N O N N N H O O O CH O CH O CH O O 3 3 H C CH 3 CH CH 3 3 3 3 6 mg (6.7 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 61) were reacted with 3 mg (8.7 µmol) of 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)- 2,2-dimethylbutanehydrazide trifluoroacetate in analogy to Intermediate 142 to give 2 mg (27% of theory) of the title nd.
HPLC (Method 12): Rt = 2.1 min; LC-MS (Method 3): Rt = 1.92 min; MS (ESIpos): m/z = 1106 (M+H)+.
Intermediate 144 N-(4-{2-[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-2,2-dimethylbutanoyl]hydrazino}oxobutyl)- N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S) (1,2-oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH N O O 3 H C H H N N 3 N N N O N N N H O O O CH O CH O CH O O CH 3 3 3 H C CH CH 3 3 3 3 To a solution of 5 mg (5.6 µmol) of arboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S) methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)(1,2-oxazinanyl)oxo phenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide in 1 ml of DMF were added 7.65 mg (22.5 µmol) of 4-(2,5-dioxo-2,5-dihydro-1H- pyrrolyl)-2,2-dimethylbutanehydrazide trifluoroacetate, 3.2 mg (16.9 µmol) of EDC, 1.96 µl (11.3 µmol) of diisopropylethylamine and 2.6 mg (16.9 µmol) of HOBT. The reaction e was stirred at RT for 3 h. Subsequently, a further 0.95 mg (2.8 µmol) of 4-(2,5-dioxo-2,5-dihydro-1H- pyrrolyl)-2,2-dimethylbutanehydrazide trifluoroacetate was added. After ng overnight, the reaction mixture was concentrated and ed by preparative HPLC. 3.5 mg (85% purity, 48% of theory) of the title compound were obtained.
LC-MS (Method 3): Rt = 1.86 min; m/z = 1094 (M+H)+.
Intermediate 145 N-[3-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)propyl]-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy 2-[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenyl cyclopropyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide H C CH H C 3 3 3 CH N O O 3 H N N N O N N N O O CH O CH O O 3 3 CH O H C CH CH 3 3 3 12 mg (14 µmol) of N-(3-aminopropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide mediate 66) were taken up in 750 µl of dioxane and admixed with 1.5 ml of saturated sodium hydrogencarbonate solution and then with 3.2 mg (21 µmol) of methyl 2,5-dioxo-2,5-dihydro-1H- pyrrolecarboxylate. The reaction mixture was stirred at RT for 1 h and then concentrated under reduced pressure. The remaining residue was purified by means of ative HPLC. After lization, 4.2 mg (32% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.94 min; MS (ESIpos): m/z = 950 (M+H)+.
Intermediate 146 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R)({(2S)[benzyl(methyl)amino]oxophenylpropan- 2-yl}amino)methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide O H C CH H3C CH 3 O 3 3 3 N O H H H N N N N N O N N N H O O O O CH O CH O O 3 CH H C CH 3 CH 3 3 3 3 9 mg (9.8 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) ({(2S)[benzyl(methyl)amino]oxophenylpropanyl}amino)methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 73) were reacted in analogy to Intermediate 133 with 10 mg (39 µmol) of 6-(2,5- dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide to give 1.8 mg (15% of theory) of the title compound.
HPLC (Method 12): Rt = 2.2 min; LC-MS (Method 9): Rt = 5.11 min; MS s): m/z = 1128 (M+H)+.
Intermediate 147 N-{4-[(2,5-dioxopyrrolidinyl)oxy]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S,3S)(benzyloxy)oxophenylbutanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C O O 3 3 3 CH O N O 3 H N O N N O N N O O O CH O CH O O 3 3 CH H C H C CH CH 3 3 3 3 3 16 mg (17 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S,3S)(benzyloxy)oxophenylbutanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 70) were dissolved in 2 ml of dichloromethane and admixed with 2.6 mg (23 mmol) of 1-hydroxypyrrolidine-2,5-dione and then with 4 mg (21 µmol) of imethylaminopropyl) ethylcarbodiimide hydrochloride. After stirring at RT for 2 h, the same amounts of 1- hydroxypyrrolidine-2,5-dione and 1-(3-dimethylaminopropyl)ethylcarbodiimide hydrochloride were added once again. Then stirring at RT ght, the reaction mixture was concentrated under reduced pressure. The remaining residue was purified by means of preparative HPLC. After lyophilization, 10 mg (56% of theory) of the title compound were ed.
HPLC (Method 5): Rt = 2.0 min; Intermediate 148 (2-{[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoyl](methyl)amino}ethyl)amino] oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl {[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidin yl}methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH 3 N O O H H H N N N N N O N N N O O O CH O CH O CH O O 3 3 3 CH H C CH3 CH 3 3 3 6 mg (7 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 61) were combined with 2.8 mg (8 µmol) of N-(2-aminoethyl)(2,5-dioxo-2,5- dihydro-1H-pyrrolyl)-N-methylbutanamide trifluoroacetate, 10.1 mg (27 µmol) of O-(7- zotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and 5 µl of N,N- diisopropylethylamine in 2 ml of DMF and stirred at RT ght. Then another 5 mg (23.5 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium uorophosphate and 3 µl of N,N- diisopropylethylamine were added. After stirring at RT for a further 5 h, the solvent was removed under reduced pressure and the remaining residue was purified by means of preparative HPLC.
The corresponding fractions were concentrated and, by lyophilization from dioxane, 1.3 mg (15% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 2.1 min; LC-MS d 2): Rt = 1.21 min; MS (ESIpos): m/z = 1120 (M+H)+.
Intermediate 149 N-{4-[(2-{[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoyl]amino}ethyl)(methyl)amino] oxobutyl}-N-methyl-L-valyl-N-[(3 R,4 S,5 S)methoxy{(2 (1 R,2R)methoxymethyl {[(1 S,2 R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidin yl}methyloxoheptanyl]-N-methyl-L-valinamide 6 mg (7 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3 R,4 S,5 S)methoxy{(2 S) [(1 R,2 R)methoxymethyl{[(1 S,2 R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 61) were combined with 3.1 mg (9 µmol) of 4-(2,5-dioxo-2,5-dihydro-1 H-pyrrol [2-(methylamino)ethyl]butanamide trifluoroacetate, 10.1 mg (27 µmol) of O-(7- azabenzotriazolyl)-N,N,N',N' -tetramethyluronium hexafluorophosphate and 5 µl of N,N - diisopropylethylamine in 2 ml of DMF, and the mixture was stirred at RT for 4 h. Then the solvent was removed under reduced pressure and the remaining residue was purified by means of preparative HPLC. The corresponding fractions were concentrated and, by lyophilization from dioxane, 1 mg (13.4% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 2.1 min; LC-MS d 1): Rt = 0.89 min; MS (ESIpos): m/z = 1121 (M+H)+.
Intermediate 150 2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3 R,4 S,5 S)methoxy{(2 S)[(1 R,2 R)methoxymethyloxo{[(1 S,2 R) phenyl(propylcarbamoyl)cyclopropyl]amino}propyl]pyrrolidinyl}methyloxoheptan yl]-N-methyl-L-valinamide O H3C CH H3C 3 CH 3 HN O O H H H N N N N N O N N N H O O O O CH3 O CH O O 3 CH H3C CH CH 3 3 3 7.9 mg (9 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyloxo{[(1S,2R)phenyl(propylcarbamoyl)cyclopropyl] amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 3 ml of DMF and then admixed with 10.4 mg (54 µmol) of 1-(3-dimethylaminopropyl) arbodiimide hydrochloride, 8.3 mg (54 µmol) of 1-hydroxy-1H-benzotriazole hydrate, 9 µl of N,N-diisopropylethylamine and with 9.5 mg (36 µmol) of commercially available 6-(2,5-dioxo- 2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was d at RT overnight and then concentrated under high vacuum. The remaining residue was purified by means of preparative HPLC. Thus, 4.3 mg (22% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 6): Rt = 1.9 min; LC-MS (Method 9): Rt = 4.93 min; MS (ESIpos): m/z = 1078 (M+H)+. ediate 151 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)carbamoylphenylcyclopropyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- namide O H C CH H C 3 3 3 CH O O 3 NH H 2 H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 The compound was prepared analogously to Intermediate 150, proceeding from the compound in Intermediate 81.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 1036 (M+H)+.
Intermediate 152 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)(ethoxycarbonyl)phenylcyclopropyl] amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]- N-methyl-L-valinamide CH 3 O H C CH H C 3 3 3 CH O O O 3 H H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 mg (12 µmol) of arboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) 2R)(ethoxycarbonyl)phenylcyclopropyl]amino}methoxymethyloxopropyl] pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 3 ml of DMF and then admixed with 8.9 mg (23 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'- tetramethyluronium hexafluorophosphate, 10 µl of N,N-diisopropylethylamine and with 12 mg (47 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide. The mixture was stirred at RT for 1 h. This was followed by concentration under high vacuum and purification of the ing residue by means of preparative HPLC. Thus, 5.8 mg (37 % of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 6): Rt = 2.0 min; LC-MS (Method 9): Rt = 4.99 min; MS (ESIpos): m/z = 1066 (M+H)+.
Intermediate 153 N-[1-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)-12,15-dioxo-3,6,9-trioxa-13,14-diazaoctadecan methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)- 1-(1,2-oxazinanyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyl- 1-oxoheptanyl]-N-methyl-L-valinamide 2012/075277 H C CH H C O 3 3 3 CH N O O H H H N N N N O O N N N O H O CH O O 3 O CH 3 O O CH H C CH H C 3 3 3 To a solution of 5 mg (5.6 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S) methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)(1,2-oxazinanyl)oxo phenylpropanyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide in 1 ml of DMF were added 9.7 mg (22.5 µmol) of 3-(2-{2-[2-(2,5-dioxo-2,5-dihydro- 1H-pyrrolyl)ethoxy]ethoxy}ethoxy)propanehydrazide trifluoroacetate, 3.2 mg (16.9 µmol) of EDC, 1.96 µl (11.3 µmol) of isopropylethylamine and 2.6 mg (16.9 µmol) of HOBT. The reaction mixture was stirred at RT for 3 h. uently, a further 1.2 mg (2.8 µmol) of 3-(2-{2-[2- ioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethoxy)propanehydrazide trifluoroacetate were added. The reaction mixture was stirred at RT overnight and then purified by preparative HPLC. 3.6 mg (51% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.90 min; m/z = 1185 (M+H)+.
Intermediate 154 (2R,3S)amino{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutan- 2-yl (3R,4S,7S,10S)[(2S)-butanyl]-7,10-diisopropyl(2-{(2S)[(1R,2R)methoxy methyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}oxoethyl)-5,11-dimethyl-6,9-dioxooxa-5,8,11-triazapentadecan- -oate O H C CH H C 3 3 3 CH N O NH O 3 H H 2 H N N N O N N O N N N H O O O O CH O CH O CH O O 3 CH 3 H C CH 3 CH 3 3 3 3 mg (17 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S)(1,2-oxazinanyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide were dissolved in ml of dichloromethane and then admixed with 12.8 mg (67 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride, 10 mg (83 µmol) of 4- dimethylaminopyridine and with 10.3 mg (33 µmol) of commercially ble benzyl N-(tert- butoxycarbonyl)-L-threoninate. The mixture was heated to reflux for 4 h. Then the same amounts of coupling reagent and 4-dimethylaminopyridine were added again and the reaction mixture was heated under reflux overnight. Subsequently, the reaction mixture was diluted with dichloromethane and ted by shaking once with water, and the organic phase was removed and concentrated under high . The ing residue was purified by means of preparative HPLC. Thus, 7.7 mg (37% of theory) of the protected intermediate were obtained as a colourless foam.
HPLC (Method 12): Rt = 2.5 min; LC-MS (Method 1): Rt = 1.13 min; MS (ESIpos): m/z = 1190 (M+H)+.
Subsequently, the benzyl ester protecting group was removed by hydrogenation under rd hydrogen pressure in methanol over 10% palladium/activated carbon, and the acid thus obtained, as described in Intermediate 151, was joined to 6-(2,5-dioxo-2,5-dihydro-1H-pyrrol yl)hexanehydrazide. In a last step, the Boc protecting group was detached with trifluoroacetic acid.
The remaining residue was ed by means of preparative HPLC. Thus, 0.22 mg (2.5% of theory over 3 stages) of the title compound was obtained as a colourless foam.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.81 min; MS (ESIpos): m/z = 1207 (M+H)+.
Intermediate 155 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)aminooxophenylpropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide O H C CH H C 3 CH O 3 O 3 3 NH H 2 H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 This compound was prepared in analogy to the synthesis described in Intermediate 152, from N-(3- carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)aminooxo phenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide and commercially available -dioxo-2,5- dihydro-1H-pyrrolyl)hexanehydrazide.
HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.82 min; MS s): m/z = 1024 (M+H)+.
Intermediate 156 N-(3-{[(1-{[(2,5-dioxopyrrolidinyl)oxy]carbonyl}cyclopropyl)carbonyl]amino}propyl)-N- methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R) (1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl- eptanyl]-N-methyl-L-valinamide O H3C CH3 H C CH N O O O 3 3 H H N N N N O O N N N H O O O CH3 O CH O O CH3 H3C CH3 3 CH This compound was prepared in analogy to the synthesis described in the last stage of Intermediate 131, from N-(3-aminopropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide and 1,1'- [cyclopropane-1,1-diylbis(carbonyloxy)]dipyrrolidine-2,5-dione, which had been obtained from the corresponding oxylic acid beforehand.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.92 min; MS (ESIpos): m/z = 1080 (M+H)+.
Intermediate 157 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH NH O O 3 H 2 H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 mg (18 µmol) of (N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyl pyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 3.8 ml of DMF and then admixed with 27 mg (70 µmol) of O-(7-azabenzotriazol yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 12 µl of N,N-diisopropylethylamine and with 14 mg (53 µmol) of commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrol yl)hexanehydrazide. The reaction mixture was stirred at RT for 1 h. This was followed by concentration under high vacuum and cation of the remaining residue by means of preparative HPLC. Thus, 6.2 mg (33% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.85 min; MS (ESIpos): m/z = 1063 (M+H)+. 1H-NMR (500 MHz, DMSO-d6, characteristic s): δ = 10.8 (d, 1H), 9.8-9.7 (m, 2H), 9.6 and 9.4 (2m, 1H), 8.9, 8.88, 8.78 and 8.75 (4d, 1H), 8.08 and 7.85 (2d, 1H), 7.6-6.9 (m, 9H), 4.7-4.4 (m, 3H), 3.4 (t, 2H), 3.23, 3.2, 3.18, 3.0, and 2.99 (5s, 9H), 2.8 (m, 3H), 2.1 (t, 2H), 1.06 and 1.01 (2d, 3H), 0.95-0.8 (m, 15H), 0.8-0.75 (dd, 3H).
Intermediate 158 N-[4-({(2R)[(2,5-dioxopyrrolidinyl)oxy]methyloxopentanyl}amino)oxobutyl]-N- methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzylamino)oxo propanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide O H3C CH H C 3 3 CH 3 HN O O H H H N N N N N O O N N O O O H C 3 O CH O CH3 O O 3 CH H C CH3 CH 3 3 3 3 WO 87716 13 mg (14.7 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)(benzylamino)oxophenylpropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were dissolved in 4 ml of dimethylformamide and then admixed with 9.4 mg (25 µmol) of O-(7- zotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 6 µl of N,N- diisopropylethylamine and with 7 mg (31 µmol) of commercially available tert-butyl D-leucinate hloride trifluoroacetate. The mixture was stirred at RT for 5 h and then concentrated under reduced re. The remaining residue was purified by means of preparative HPLC. After lyophilization from dioxane/water, 6.5 mg (49% of theory) of the protected intermediate were obtained as a colourless foam.
HPLC (Method 5): Rt = 2.2 min; LC-MS (Method 1): Rt = 1.21 min; MS (ESIpos): m/z = 1076 (M+H)+.
Trifluoroacetic acid in dichloromethane was first used to detach the Boc protecting group from this protected intermediate, giving 6.2 mg (99% of theory) of the ected compound. 5.2 mg (5 µmol) of this intermediate were taken up in 1.5 ml of dichloromethane and reacted with 0.8 mg (7 µmol) of N-hydroxysuccinimide in the presence of 1.2 mg (6 µmol) of 1-(3-dimethylaminopropyl)- 3-ethylcarbodiimide hydrochloride and 0.16 mg (1 µmol) of thylaminopyridine. After stirring at RT for 2 h, the reaction mixture was concentrated and purified by means of preparative HPLC. 1.3 mg of the title compound were obtained, some of which was hydrolysed to the reactant.
Intermediate 159 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzylamino)oxophenylpropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide H C CH H3C 3 3 CH H O O 3 N N H H N N N N O N N N O H O O O CH3 O CH3 O O CH H3C CH CH3 3 This nd was prepared in analogy to the synthesis described in Intermediate 157, from N-(3- ypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzylamino) oxophenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide and commercially available 6-(2,5-dioxo-2,5- dihydro-1H-pyrrolyl)hexanehydrazide. 2012/075277 Yield: 6 mg (53% of theory) HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.94 min; MS (ESIpos): m/z = 1114 .
Intermediate 160 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzylamino)(1H-indolyl)oxopropan- 2-yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH NH O O 3 N H H N N N N O N N N O H O O O CH O CH O O 3 3 H C H C CH CH 3 3 3 3 HN This compound was prepared in analogy to the synthesis described in Intermediate 157, from 20 mg (21 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)(benzylamino)(1H-indolyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide and commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide.
Yield: 13 mg (52% of theory) HPLC (Method 5): Rt = 1.9 min; LC-MS d 1): Rt = 0.92 min; MS (ESIpos): m/z = 1153 (M+H)+.
Intermediate 161 N-(6-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxohexyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH O 3 NH2 O H N H H N N N N O N N N O H O O O CH O CH O O 3 3 H C H C CH3 CH 3 3 3 This compound was prepared in analogy to the sis described in Intermediate 157, from N-(5- carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indol- 3-yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide and commercially available 6-(2,5-dioxo-2,5- dihydro-1H-pyrrolyl)hexanehydrazide.
Yield: 0.8 mg (16% of theory) HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.78 min; MS s): m/z = 1092 .
Intermediate 162 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl) phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide H C CH H C 3 3 3 CH 3 N O O H H N O N N O N N N O O O CH O CH O O 3 3 CH H C CH CH 3 O 3 3 3 18 mg (20 µmol) of N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl] amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 64) were dissolved in 3.2 ml of dichloromethane and admixed with 22 mg (190 mmol) of 1-hydroxypyrrolidine-2,5-dione and then with 11 mg (60 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride and 0.24 mg (0.17 µmol) of DMAP.
After stirring at RT for 2 h, another 22 mg (190 mmol) of 1-hydroxypyrrolidine-2,5-dione, 11 mg WO 87716 2012/075277 (60 µmol) of 1-(3-dimethylaminopropyl)ethylcarbodiimide hydrochloride and 0.24 mg (0.17 µmol) of DMAP were added and the reaction mixture was stirred at RT for a further hour. This was followed by concentration under reduced pressure. The remaining residue was purified by means of preparative HPLC. After lyophilization, 8.2 mg (41% of ) of the title compound were obtained.
HPLC (Method 5): Rt = 2.0 min; LC-MS d 11): Rt = 0.9 min; MS (ESIpos): m/z = 1024 (M+H)+.
Intermediate 163 [(1S,2R)aminophenylcyclopropyl](1,4-dihydro-3H-2,3-benzoxazinyl)methanone trifluoroacetate CF3COOH x H N 2 O First, proceeding from 265 mg (0.82 mmol) of tert-butyl (1S,2R)(hydroxycarbamoyl) phenylcyclopropyl carbamate (Starting Compound 7), by reaction with 1,2- bis(bromomethyl)benzene, analogously to a literature method (see H. King, J. Chem. Soc. 1942, 432), the Boc-protected tert-butyl (1S,2R)(1,4-dihydro-3H-2,3-benzoxazinylcarbonyl) phenylcyclopropyl carbamate intermediate was prepared.
Yield: 108 mg (34% of ) LC-MS (Method 2): Rt = 1.3 min; MS (ESIpos): m/z = 395 (M+H)+. 108 mg (0.27 mmol) of this intermediate were taken up in 3.7 ml of dichloromethane, 1.8 ml of trifluoroacetic acid were added, and the mixture was stirred at RT for 15 min. This was followed by concentration under reduced pressure and lyophilization of the remaining residue from dioxane. 112 mg of the title compound were obtained in tative yield as a colourless foam.
WO 87716 LC-MS (Method 1): Rt = 0.7 min; MS (ESIpos): m/z = 295 (M+H)+.
Intermediate 164 N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)(1,4-dihydro-3H-2,3- benzoxazinylcarbonyl)phenylcyclopropyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C CH 3 3 3 3 N O H H N N N O HN N O O CH O CH O O H C 3 3 3 H C CH CH 3 3 3 x CF3COOH 166 mg (0.196 mmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(3R,4S,5S)- 1-{(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide (Intermediate 10) were taken up in 40 ml of DMF and admixed successively with 80 mg (0.196 mmol) of R)aminophenylcyclopropyl](1,4- o-3H-2,3-benzoxazinyl)methanone trifluoroacetate (Intermediate 163), 112 mg (0.294 mmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 682 µl (3.9 mmol) of N,N-diisopropylethylamine. The mixture was uently d at RT overnight. The reaction mixture was then concentrated under reduced pressure, the residue was taken up in ethyl acetate and the solution was washed with saturated aqueous sodium chloride solution. The organic phase was dried over magnesium sulphate, filtered and concentrated. The residue was finally ed by preparative HPLC. In this way, 19 mg (9% of theory) of the Fmocprotected intermediate N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R){[(1S,2R)(1,4-dihydro-3H-2,3-benzoxazinylcarbonyl) phenylcyclopropyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide were obtained.
HPLC (Method 5): Rt = 1.68 min; LC-MS (Method 1): Rt = 1.51 min; MS (ESIpos): m/z = 1083 (M+H)+. 19 mg (0.015 mmol) of this ediate were dissolved in 4 ml of DMF. After 817 µl of piperidine had been added, the reaction mixture was stirred at RT for 5 min. This was followed by concentration under reduced pressure, and the residue was first digested with diethyl ether and then purified by means of preparative HPLC (eluent: itrile + 0.1% TFA / 0.1% aq. TFA).
The corresponding fractions were combined, the solvent was removed under reduced re and then the residue was lyophilized from dioxane/water. 12 mg (92% of theory) of the title compound were obtained as a colourless foam.
HPLC (Method 6): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.94 min; MS (ESIpos): m/z = 861 (M+H)+.
Intermediate 165 N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R)(1,4- dihydro-3H-2,3-benzoxazinylcarbonyl)phenylcyclopropyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H3C CH H C CH 3 3 N O 3 H N H N N N 2 O N N O O CH O CH O O H C 3 3 3 H3C CH CH 3 3 20 mg (0.021 mmol) of ediate 164 were used, in y to the preparation of Intermediate 97, with benzyl 3-oxohexyl carbamate in the presence of sodium cyanoborohydride and subsequent hydrogenolytic detachment of the Z protecting group (with 5% palladium on charcoal as a catalyst, in methanol as a solvent), to prepare the title compound.
Yield: 4.5 mg (23% of theory over 2 stages) HPLC (Method 12): Rt = 1.9 min; LC-MS d 1): Rt = 0.9 min; MS (ESIpos): m/z = 960 (M+H)+.
Intermediate 166 N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(1S,2R)(1,4-dihydro-3H-2,3-benzoxazinylcarbonyl) phenylcyclopropyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy oxoheptanyl]-N-methyl-L-valinamide O O H C CH H C 3 3 3 CH 3 N O H H N N N N O N N O O O CH O CH O O H C 3 3 3 H C CH CH 3 3 3 4.4 mg (4.5 µmol) of Intermediate 165 were taken up in 1 ml of 1:1 dioxane/water and then admixed with 1 mg (6.8 µmol) of methyl 2,5-dioxo-2,5-dihydro-1H-pyrrolecarboxylate and with 50 µl of saturated aqueous sodium hydrogencarbonate solution. The reaction mixture was stirred at RT for 30 min. Then another 50 µl of the saturated aqueous sodium hydrogencarbonate on were added and the on mixture was stirred at RT for a further 15 min and then concentrated under reduced pressure. The remaining residue was purified by means of preparative HPLC. After lyophilization, 1 mg (21% of theory) of the title compound were obtained as a colourless foam.
HPLC d 12): Rt = 2.1 min; LC-MS (Method 1): Rt = 1.08 min; MS (ESIpos): m/z = 1040 (M+H)+.
Intermediate 167 benzyl 3-{2-[2-(2-oxoethoxy)ethoxy]ethoxy}propanoate O H O O The title compound was prepared from 6 g (21.55 mmol) of commercially available 3-{2-[2-(2- hydroxyethoxy)ethoxy]ethoxy}propanoic acid under standard ions, first by esterification with benzyl chloride and caesium carbonate and subsequent oxidation with sulphur trioxidepyridine complex.
Yield: 611 mg (10% of theory over 2 stages) LC-MS (Method 2): Rt = 1.69 min; MS (ESIpos): m/z = 311 (M+H)+.
Intermediate 168 N-(2-{2-[2-(2-carboxyethoxy)ethoxy]ethoxy}ethyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyl pyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH NH O 3 H 2 H N HO O O N N O O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 First, in analogy to the synthesis bed in Intermediate 69, by coupling of N-[(9H-fluoren ylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)carboxymethoxymethylhexanyl]- N-methyl-L-valinamide (Intermediate 4) and Nα-{(2R,3R)methoxymethyl[(2S)-pyrrolidin- ropanoyl}-L-tryptophanamide trifluoroacetate (Intermediate 49) in the presence of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and uent detachment of the Fmoc protecting group by means of piperidine, the amine compound N-methyl- L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide was prepared as the trifluoroacetate. 25 mg (0.028 mmol) of this compound and 17.5 mg (0.06 mmol) of Intermediate 167 were combined in 2 ml of methanol and admixed with 12.6 mg (0.14 mmol) of borane-pyridine complex and 2.5 ml of acetic acid. The reaction mixture was stirred at RT ght. Then the same amounts of borane-pyridine complex and acetic acid were added once again and the reaction mixture was stirred at RT for a further 24 h. This was ed by concentration under d pressure, and the e was purified by means of preparative HPLC. After concentration of the corresponding fractions and lyophilization from 1:1 dioxane/water, 26.5 mg (88% of theory) of the Z-protected title compound were obtained.
HPLC (Method 12): Rt = 2.04 min; LC-MS (Method 1): Rt = 0.97 min; MS s): m/z = 1064 (M+H)+. mg (0.024 mmol) of this intermediate were taken up in 10 ml of methanol and hydrogenated over 10% palladium on ted carbon under standard hydrogen pressure at RT for 45 min. The st was then filtered off and the t was removed under reduced pressure. After lyophilization from e, 19.7 mg (85% of theory) of the title compound were obtained.
HPLC d 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.83 min; MS (ESIpos): m/z = 974 (M+H)+.
Intermediate 169 N-{2-[2-(2-{3-[(2,5-dioxopyrrolidinyl)oxy]oxopropoxy}ethoxy)ethoxy]ethyl}-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH NH O O 3 H N O O O N N O N O N N O O O CH O CH O O 3 3 CH O H C CH CH 3 NH 3 3 3 mg (10 µmol) of Intermediate 168 were dissolved in 3 ml of DMF and admixed with 3.5 mg (30 mmol) of 1-hydroxypyrrolidine-2,5-dione and then with 2.4 mg (10 µmol) of 1-(3- dimethylaminopropyl)ethylcarbodiimide hydrochloride and 5 µl of N,N-diisopropylethylamine.
After stirring at RT for 20 h, 8 mg (0.02 mmol) of HATU were added and the reaction mixture was stirred once again at RT overnight and then concentrated under reduced pressure. The remaining residue was purified by means of preparative HPLC. After lyophilization from dioxane, 8.6 mg (64% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 11): Rt = 0.81 min; MS (ESIpos): m/z = 1071 (M+H)+.
Intermediate 170 N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(2S,3S)(1,2-oxazinanyl)oxophenylbutanyl]amino} oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H3C 3 3 CH O 3 N H N H N N N O 2 N N O O CH O CH O O 3 H C 3 CH 3 H C H C CH 3 3 3 3 This compound was prepared in analogy to Intermediate 101 over 2 stages, proceeding from 26 mg (0.028 mmol) of Intermediate 15.
Yield: 16.7 mg (63% of theory over 2 stages) HPLC (Method 12): Rt = 1.9 min; LC-MS d 1): Rt = 0.81 min; MS (ESIpos): m/z = 914 (M+H)+.
Intermediate 171 N-(6-{[4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoyl]amino}hexyl)-N-methyl-L-valyl-N- [(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S,3S)(1,2-oxazinan yl)oxophenylbutanyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N- methyl-L-valinamide H C CH H C 3 3 3 CH N O 3 O H H H N N N N O N N N O O O CH O CH O O H C 3 3 3 H C O H C CH CH 3 3 3 3 6.7 mg (7.3 µmol) of the compound formed from Intermediate 170 and 3 mg (14.7 µmol) of commercially available 4-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)butanoic acid were taken up in 2 ml of DMF and admixed with 5.6 mg (14.7 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'- tetramethyluronium uorophosphate (HATU) and 2 µl of N,N-diisopropylethylamine. The mixture was stirred at RT for 30 min. The reaction mixture was concentrated and the residue was purified by means of preparative HPLC. The corresponding fractions were combined, the solvent was removed under reduced re and then the residue was lyophilized from dioxane. Thus, 4.5 mg (56% of theory) of the title compound were ed.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 1.12 min; MS (ESIpos): m/z = 1079 (M+H)+.
Intermediate 172 benzyl 2-{2-[2-(2-oxoethoxy)ethoxy]ethoxy}ethyl carbamate O N O H O O O O The title compound was prepared from commercially ble 2-(2- aminoethoxy)ethoxy]ethoxy}ethanol under standard ions, by first introducing the Z protecting group and then oxidizing with sulphur trioxide-pyridine complex.
HPLC (Method 12): Rt = 1.4 min; LC-MS (Method 11): Rt = 0.65 min; MS (ESIpos): m/z = 326 (M+H)+.
Intermediate 173 benzyl {2-[2-(2-oxoethoxy)ethoxy]ethyl carbamate O N O O H The title compound was prepared analogously to Intermediate 172 from commercially available 2- [2-(2-aminoethoxy)ethoxy]ethanol under standard conditions, by first introducing the Z ting group and then oxidizing with sulphur trioxide-pyridine complex.
HPLC d 12): Rt = 1.3 min; LC-MS (Method 11): Rt = 0.68 min; MS (ESIpos): m/z = 282 (M+H)+.
Intermediate 174 N-(2-{2-[2-(2-aminoethoxy)ethoxy]ethoxy}ethyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenyl 2012/075277 cyclopropyl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide H C CH H C CH 3 3 3 3 N O H H N O O N N O H N O N N 2 O O CH O CH O O H C 3 3 CH 3 H C CH 3 3 3 47 mg (0.05 mmol) of Intermediate 16 were reductively aminated in analogy to the preparation of Intermediate 167 with benzyl 2-{2-[2-(2-oxoethoxy)ethoxy]ethoxy}ethyl carbamate in the presence of borane-pyridine complex. Subsequently, the Z protecting group was removed by hydrogenolytic means with 5% palladium on charcoal as a catalyst and in ol as a solvent, and 38 mg (66% of theory over 2 stages) of the title nd were prepared.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.8 min; MS (ESIpos): m/z = 988 (M+H)+.
Intermediate 175 N-[2-(2-{2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethoxy)ethyl]-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2- oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH 3 N O O H H N O O N N O N O N N O O CH O CH O O H C 3 3 3 O H C CH CH 3 3 3 The preparation was effected in y zu Intermediate 166, proceeding from 34 mg (0.03 mmol) of Intermediate 174.
Yield: 8.3 mg (23% of theory) HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.97 min; MS (ESIpos): m/z = 1068 (M+H)+.
Intermediate 176 N-(2-{2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethyl)-N-methyl-L-valyl-N- [(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- namide O H C CH H C CH 3 3 3 3 NH O H 2 H N N O N N O O N N O O O CH O CH O O H C 3 3 CH 3 H C CH 3 3 3 The preparation was effected in analogy to Intermediates 174 and 175, commencing with the reductive amination of Intermediate 16 with ediate 173, subsequent deprotection and formation of the maleimide.
HPLC (Method 12): Rt = 1.8 min; LC-MS (Method 11): Rt = 0.8 min; MS (ESIpos): m/z = 981 (M+H)+.
Intermediate 177 N-[2-(2-{2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethoxy)ethyl]-N-methyl-L- N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino(1H-indolyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan methyl-L-valinamide H C CH H C CH 3 3 3 3 NH O O H H N O O N N O N O N N O O CH O CH O O H C 3 3 O H C CH CH 3 NH 3 3 3 The preparation was effected in analogy to Intermediates 174 and 175, commencing with the reductive amination of Intermediate 16 with Intermediate 172, subsequent deprotection and formation of the maleimide.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 1025 (M+H)+. ediate 178 N-{4-[(2,5-dioxopyrrolidinyl)oxy]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyl pyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 CH 3 NH 3 3 H 2 O O H N O N N O N N N O O O CH O CH O O CH 3 3 CH 3 O H C CH 3 3 3 The preparation was effected in y to Intermediates 162, proceeding from 6 mg of ediate 82.
LC-MS (Method 1): Rt = 0.82 min; MS (ESIpos): m/z = 953 (M+H)+.
Intermediate 179 4-[(1E,3S)aminophenylbutenyl]benzenesulphonic acid trifluoroacetate x CF3COOH A mixture of 13.6 mg (0.06 mmol) of palladium(II) acetate, 469 mg (1.46 mmol) of potassium 4- iodobenzenesulphonate, 300 mg (1.21 mmol) of (S)-tert-butyl 1-phenylbutenyl carbamate, 16.5 mg (0.12 mmol) of phenylurea and 167.6 mg (1.21 mmol) of potassium carbonate in 7.5 ml of DMF was heated to 160°C in a microwave for 15 min. The crude product was subsequently purified directly by preparative HPLC. This gave 312 mg of a e of 31% of the BOC- protected compound and 69% of the free amine.
This mixture was subsequently taken up in 30 ml of dichloromethane, admixed with 1 ml of trifluoroacetic acid and stirred at RT for 20 h. After concentrating under reduced pressure, the residue was stirred with diethyl ether, and the itate formed was filtered off with suction and washed with diethyl ether. This gave 200 mg (62% of theory) of the title nd.
LC-MS (Method 11): Rt = 0.44 min; MS (ESIpos): m/z = 304 (M+H)+.
Intermediate 180 )aminophenylbutyl]benzenesulphonic acid x CF3COOH 100 mg (0.25 mmol) of 4-[(1E,3S)aminophenylbutenyl]benzenesulphonic acid trifluoroacetate were suspended in 10 ml of acetic acid and a few drops of DMF and water, admixed with 70 mg (0.07 mmol) of palladium on charcoal (10%) and hydrogenated at hydrogen pressure 2.2 bar for 24 h. The solution was filtered and the filtrate purified by prep. HPLC. 29 mg (76% purity, 21% of theory) of t were obtained.
LC-MS (Method 1): Rt = 0.46 min; MS (ESIpos): m/z = 306 (M+H)+.
Intermediate 181 N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyloxo{[(2S,3E)phenyl(4-sulphophenyl)buten yl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH CH3 O 3 3 O 3 3 H H C H 3 N N N H C O N N 3 O O CH3 O CH O O CH 3 3 H C CH 3 3 CH O O To a solution of 90 mg (0.13 mmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S)- 1-{(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide in 4 ml of DMF were added 60 mg (0.16 mmol) of HATU and 69 µl of (0.39 mmol) Hünig's base. The reaction mixture was stirred at RT for 30 min and then admixed with 60 mg (0.15 mmol) 60.3 mg (0.13 mmol) of 4-[(1E,3S)aminophenylbuten zenesulphonic acid trifluoroacetate. After ng overnight, the reaction mixture was purified by prep. HPLC. This gave 127 mg of a 44:56 mixture of the title compound and of the already deprotected amine.
LC-MS (Method 1): Rt = 1.21 min; MS (ESIpos): m/z = 971 (M+H)+; R t = 0.84 min; MS (ESIpos): m/z = 871 (M+H)+ for the deprotected compound.
Intermediate 182 N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo {[(2S,3E)phenyl(4-sulphophenyl)butenyl]amino}propyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H3C CH H C 3 O 3 CH 3 H H N N N HN N O O CH O CH3 O O CH 3 H C CH 3 3 CH CF3COOH x O O OH 90 mg of Intermediate 180 were dissolved in 4.6 ml of dichloromethane, and 0.92 ml of oroacetic acid was added. The reaction mixture was stirred at RT for 30 min and then concentrated. The crude t obtained was purified by prep. HPLC. 91 mg (98% of theory) of the target nd were obtained.
LC-MS (Method 1): Rt = 0.85 min; MS (ESIpos): m/z = 871 (M+H)+ Intermediate 183 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy- 2-methyloxo{[(2S,3E)phenyl(4-sulphophenyl)butenyl]amino}propyl]pyrrolidin yl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH3 H3C 3 CH O 3 H H N HO N N N N O O O CH O CH O O CH 3 C CH3 3 3 H3 CH O O OH 16.7 µl (0.03 mmol) of a 15% aqueous succinaldehyde solution were initially charged in 943 µl of methanol and admixed with 17 mg (0.02 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyloxo{[(2S,3E)phenyl(4-sulphophenyl)buten yl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate (Intermediate 181) and 1.1 µl (0.02 mmol) of acetic acid. The reaction mixture was stirred for 5 min at RT and then 2.9 µl (0.02 mmol) of borane-pyridine complex were added. After 1 h, a further 2 lents each of succinaldehyde, acetic acid and borane-pyridine x were added and the mixture was stirred at RT for 20 h. The reaction mixture was then purified by prep.
HPLC.
This gave 20 mg (83% , 80% of ) of the title compound.
LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 957 (M+H)+ Intermediate 184 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo{[(2S,3E) phenyl(4-sulphophenyl)butenyl]amino}propyl]pyrrolidinyl}methyloxoheptan yl]-N-methyl-L-valinamide O H C CH H C CH O 3 3 O 3 3 H H H N N N N N N N N H O O O O CH O CH O O CH 3 3 H C CH 3 3 3 CH O O 8 mg (7.5 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyloxo{[(2S,3E)phenyl(4-sulphophenyl)buten yl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide, 2.8 mg (8.2 µmol) of 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide trifluoroacetate, 3.4 mg (9 µmol) of HATU and 3.9 µl of s base were stirred in 0.77 ml of DMF at RT for 20 h.
Subsequently, the reaction mixture was purified by prep. HPLC. 3 mg (31% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.90 min; MS (ESIpos): m/z = 1164 (M+H)+ ediate 185 N-{4-[(2,5-dioxopyrrolidinyl)oxy]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyloxo{[(2S,3E)phenyl(4-sulphophenyl)buten yl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 CH O 3 3 H H N N O N N N O O O CH O CH O O CH O 3 H C CH 3 3 3 3 CH O O To a solution of 8 mg (7.5 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S) methoxy{(2S)[(1R,2R)methoxymethyloxo{[(2S,3E)phenyl(4- phenyl)butenyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl- L-valinamide in 2 ml of DMF were added 8.6 mg (74.8 µmol) of N-hydroxysuccinimide, 8.5 mg (22.4 µmol) of EDCI and 0.1 mg (0.75 µmol) of DMAP. The reaction mixture was stirred at RT for 20 h. Subsequently, 1.3 µl (7.5 µmol) of Hünig's base were added and the mixture was stirred for 1 h. The reaction e was then purified by prep. HPLC. 2.6 mg (72% purity, 21% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.89 min; MS (ESIpos): m/z = 1054 (M+H)+ Intermediate 186 N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyloxo{[(2R)phenyl(4-sulphophenyl)butan yl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C O 3 3 CH CH O 3 3 H C 3 H 3 N N N H C O N N 3 O O CH O CH O CH 3 O 3 3 H C CH 3 3 CH O O To a solution of 43 mg (0.06 mmol) of N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S)- 1-{(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide in 1.9 ml of DMF were added 29 mg (0.07 mmol) of HATU and 33 µl (0.19 mmol) of Hünig's base. The reaction mixture was stirred at RT for 30 min and then admixed with 29 mg (0.07 mmol) of 4-[(3R)aminophenylbutyl]benzenesulphonic acid oroacetate. After stirring overnight, the reaction e was purified by prep. HPLC.
This gave 58 mg of a 45:55 mixture of the title compound and of the already ected amine.
LC-MS (Method 1): Rt = 1.09 min; MS (ESIpos): m/z = 973 (M+H)+; R t = 0.87 min; MS s): m/z = 873 (M+H)+ for the deprotected compound.
Intermediate 187 N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo {[(2R)phenyl(4-sulphophenyl)butanyl]amino}propyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C 3 3 3 CH O 3 H H N N N CF3COOH x HN N O O CH O CH 3 O O CH 3 H C CH 3 3 CH O O 58 mg of Intermediate 186 were dissolved in 4.1 ml of dichloromethane, 0.41 ml of trifluoroacetic acid was added and the mixture was stirred at RT for 30 min. After concentration under reduced pressure, the crude product was purified by prep. HPLC. 50 mg (90% purity, 85% of theory) of the title compound were ed.
LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 873 (M+H)+ Intermediate 188 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy- 2-methyloxo{[(2R)phenyl(4-sulphophenyl)butanyl]amino}propyl]pyrrolidinyl} methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH O 3 H H N HO N N N N O O O CH O CH O O CH 3 3 3 H C CH 3 3 CH O O 171 µl (0.26 mmol) of a 15% aqueous succinaldehyde solution were initially charged in 2.5 ml of methanol and admixed with 50 mg (0.05 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyloxo{[(2R)phenyl(4-sulphophenyl)butan yl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide oroacetate and 11.6 µl (0.2 mmol) of acetic acid. The on mixture was stirred for 5 min at RT and then 30 µl (0.24 mmol) of borane-pyridine complex were added. After stirring for 24 hours, a further equivalent of borane-pyridine complex was added and the mixture was stirred for a further 2 h. The on mixture was then purified by prep. HPLC. 40 mg (90% purity, 66% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.91 min; MS (ESIpos): m/z = 959 (M+H)+ Intermediate 189 2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo{[(2R) phenyl(4-sulphophenyl)butanyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]- N-methyl-L-valinamide O H C CH3 H C O 3 3 CH3 O H H H N N N N N N N N H O O O O CH O CH O O CH 3 H3C CH 3 3 CH O O mg (9.3 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyloxo{[(2R)phenyl(4-sulphophenyl)butan no}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide, 3.5 mg (10.3 µmol) of 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide trifluoroacetate, 4.3 mg (11.2 µmol) of HATU and 4.9 µl (28 µmol) of Hünig's base were stirred in 1 ml of DMF at RT for h. Subsequently, the on mixture was purified by prep. HPLC. 4.2 mg (92% purity, 33% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.91 min; MS (ESIpos): m/z = 1166 (M+H)+ Intermediate 190 N-{4-[(2,5-dioxopyrrolidinyl)oxy]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyloxo{[(2R)phenyl(4-sulphophenyl)butan yl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 CH O 3 H H N O N N N N N O O O CH O CH O O CH O H C CH 3 3 3 CH O O To a solution of 10 mg (9.3 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S) methoxy{(2S)[(1R,2R)methoxymethyloxo{[(2R)phenyl(4- sulphophenyl)butanyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide in 2.5 ml of DMF were added 10.7 mg (93 µmol) of N-hydroxysuccinimide, 10.6 mg (28 µmol) of EDCI and 0.12 mg (0.9 µmol) of DMAP. The reaction mixture was stirred at RT for h and then purified by prep. HPLC. 3.8 mg (72% purity, 25% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.90 min; MS (ESIpos): m/z = 1055 (M+H)+ Intermediate 191 (2R,3R)-N-[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]methoxymethyl- 3-[(2S)-pyrrolidinyl]propanamide oroacetate CH N 3 H CF3COOH x N O O O The title compound was prepared in analogy to the synthesis of Intermediate 7 over two stages from Starting nd 1 and -amino(1H-indolyl)(1,2-oxazinanyl)propanone trifluoroacetate (Intermediate 99).
Yield over 2 stages: 62 mg (67% of theory) WO 87716 HPLC d 6): Rt = 1.65 min; LC-MS (Method 1): Rt = 0.7 min; MS (ESIpos): m/z = 443 (M+H)+.
Intermediate 192 N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinan- 2-yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 O 3 N 3 H H N N N O HN N O O CH O CH O O H C 3 3 H C CH CH 3 3 3 3 1015 mg (1.59 mmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)- oxymethoxymethylhexanyl]-N-methyl-L-valinamide mediate 4) were taken up in 50 ml of DMF, admixed with 654 mg (2.39 mmol) of 2-bromoethylpyridinium tetrafluoroborate (BEP) and 2.8 ml of N,N-diisopropylethylamine, and stirred at RT for 10 min.
Then 1083 mg (1.75 mmol) of (2R,3R)-N-[(2S)(1H-indolyl)(1,2-oxazinanyl) oxopropanyl]methoxymethyl[(2S)-pyrrolidinyl]propanamide trifluoroacetate (Intermediate 191) were added and then the mixture was treated in an ultrasound bath at RT for 30 min. The reaction mixture was then concentrated under reduced pressure and the e was taken up in 300 ml of ethyl acetate. The organic phase was washed successively with 5% s citric acid solution and 5% aqueous sodium hydrogencarbonate solution, dried over magnesium sulphate, filtered and concentrated. The crude product thus obtained (1684 mg), without further purification, was taken up in 20 ml of acetonitrile, 2 ml of piperidine were added and the reaction mixture was then stirred at RT for 10 min. Then the mixture was concentrated under reduced pressure and the residue was admixed with diethyl ether. The solvent was concentrated by evaporation again and the residue was purified by flash chromatography on silica gel (eluent: :1:0.1 -> 15:2:0.2 dichloromethane/methanol/17% a queous ammonia solution). The corresponding ons were combined, the solvent was d under reduced pressure and the residue was lyophilized from acetonitrile/water. Thus, 895 mg (67% over 2 stages) of the title compound were obtained.
HPLC (Method 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.84 min; MS (ESIpos): m/z = 840 . 1H NMR (500 MHz, DMSO-d 6): δ = 10.8 (d, 1H), 8.3 and 8.05 (2d, 1H), 8.0 (d, 1H), 7.5 (m, 1H), 7.3 (m, 1H), 7.15 and 7.08 (2s, 1H) 7.05-6.9 (m, 2H), 5.12 and 4.95 (2m, 1H), 4.65 (m, 1H), 4.55 (m, 1H), 4.1-3.8 (m, 4H), 3.75 (d, 1H), 3.23, 3.18, 3.17, 3.12, 2.95 and 2.88 (6s, 9H), 3.1-3.0 and 2.85 (2m, 2H), 2.65 (d, 1H), 2.4-2.2 (m, 3H), 2.15 (m, 3H), 1.95 (br. m, 2H), 1.85-0.8 (br. m, 11H), 1.08 and 1.04 (2d, 3H), 0.9-0.75 (m, 15H), 0.75-0.65 (dd, 3H) [further signals hidden under H2O peak].
Intermediate 193 N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indol yl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidin- 1-yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 O 3 3 N H N HO N N O N N O O O CH O CH O O H C 3 3 3 H C CH CH 3 3 3 50 mg (0.052 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H- indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 192) and 204 µl einer of a 15% aqueous on of 4-oxobutanoic acid were combined in 2 ml of methanol and admixed with 23.4 mg (0.252 mmol) of borane-pyridine complex and 6 µl of acetic acid. The on mixture was stirred at RT overnight. This was followed by concentration under reduced re, and the residue was purified by means of preparative HPLC. After concentration of the corresponding fractions, 38 mg (78% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 9): Rt = 4.7 min; MS (ESIpos): m/z = 926 (M+H)+.
Intermediate 194 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide O O H C CH H C 3 3 3 CH3 N O O H N H H N N N N O N N N O H O O O CH O CH3 O O 3 H C H3C CH CH 3 3 3 HN This compound was prepared in analogy to the synthesis described in Intermediate 157 from 10 mg (11 µmol) of N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)- 3-(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide and commercially available 6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanehydrazide.
Yield: 4.4 mg (35% of theory) HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.90 min; MS (ESIpos): m/z = 1133 . ediate 195 2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyl{[(2S,3S)(1,2-oxazinanyl)oxophenylbutan yl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 3 3 CH N O 3 H H N N N N O N N O O O CH O CH O O 3 H C 3 H C H C CH CH3 3 3 3 This compound was prepared in analogy to Intermediate 166, proceeding from 9 mg (0.010 mmol) of Intermediate 170.
Yield: 1.1 mg (10% of theory) HPLC (Method 12): Rt = 2.0 min; 2012/075277 LC-MS (Method 1): Rt = 0.99 min; MS (ESIpos): m/z = 994 (M+H)+.
Intermediate 196 -amino(2-oxaazabicyclo[2.2.2]octenyl)phenylpropanone trifluoroacetate CF3COOH x H N 2 O 41 mg (0.37 mmol) of 2,5-dioxopyrrolidinyl N-(tert-butoxycarbonyl)-L-phenylalaninate were taken up in 10 ml of DMF and admixed with 149 mg (0.41 mmol) of 2-oxaazabicyclo[2.2.2]oct- -ene (Starting Compound 6) and 72 µl (0.41 mmol) of N,N-diisopropylethylamine. The mixture was stirred at RT for 1 h. The solvent was removed under reduced pressure, and the residue was taken up in ethyl acetate and extracted by shaking with 5% aqueous citric acid solution and then with 5% aqueous sodium hydrogencarbonate solution. The organic phase was concentrated and the residue was ed by flash chromatography on silica gel with 10:1 toluene/ethanol as the eluent.
The corresponding fractions were ed and the solvent was removed under reduced pressure.
After the residue had been dried under high vacuum, 69 mg (47% of theory) of the Boc-protected intermediate tert-butyl (2S)(2-oxaazabicyclo[2.2.2]octenyl)oxophenylpropanyl carbamate were thus obtained as a reomer mixture.
LC-MS (Method 1): Rt = 1.1 min; MS s): m/z = 359 (M+H)+. 64 mg (0.18 mmol) of this intermediate were taken up in 10 ml of dichloromethane, 1 ml of trifluoroacetic acid was added, and the mixture was stirred at RT for 30 min. This was followed by concentration under reduced pressure and lyophilization of the remaining residue from water/dioxane. In this way, 66 mg (quant.) of the title compound were obtained as a foam.
HPLC (Method 6): Rt = 1.45 min; LC-MS (Method 3): Rt = 1.12 min; MS (ESIpos): m/z = 259 (M+H)+. ediate 197 (2R,3R)methoxymethyl-N-[(2S)(2-oxaazabicyclo[2.2.2]octenyl)oxo phenylpropanyl][(2S)-pyrrolidinyl]propanamide trifluoroacetate CH N CF3COOH x N N O O O First, (2R,3R)[(2S)(tert-butoxycarbonyl)pyrrolidinyl]methoxymethylpropanoic acid (Starting Compound 1) was released from 83 mg (0.18 mmol) of its dicyclohexylamine salt by taking it up in ethyl acetate and extractive shaking with 5% aqueous potassium hydrogensulphate solution. The organic phase was dried over magnesium sulphate, filtered and trated. The e was taken up in 10 ml of DMF and d successively with 66 mg (0.18 mmol) of (2S)- 2-amino(2-oxaazabicyclo[2.2.2]octenyl)phenylpropanone trifluoroacetate (Intermediate 196), 101 mg (0.266 mmol) of O-(7-azabenzotriazolyl)-N,N,N',N'- tetramethyluronium hexafluorophosphate (HATU) and 93 µl (0.53 mmol) of N,N- diisopropylethylamine. The mixture was stirred at RT for 30 min. The reaction mixture was then trated and the residue was purified by preparative HPLC. This gave 52 mg (56% of theory) of the Boc-protected intermediate tert-butyl (2S)[(1R,2R)methoxymethyl{[(2S)(2- oxaazabicyclo[2.2.2]octenyl)oxophenylpropanyl]amino} oxopropyl]pyrrolidinecarboxylate.
HPLC (Method 6): Rt = 2.13 min; LC-MS d 1): Rt = 1.13 min; MS (ESIpos): m/z = 528 (M+H)+. 52 mg (0.1 mmol) of this intermediate were taken up in 10 ml of dichloromethane, 1 ml of trifluoroacetic acid was added, and the mixture was stirred at RT for 20 min. This was followed by concentration under reduced pressure and stirring of the ing residue with 20 ml of diethyl ether. After 10 min, the mixture was filtered and the filter residue was dried under high vacuum. In this way, 39 mg (72% of theory) of the title compound were obtained.
HPLC (Method 6): Rt = 1.62 min; LC-MS (Method 1): Rt = 0.68 min; MS (ESIpos): m/z = 428 (M+H)+.
Intermediate 198 N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)(2- oxaazabicyclo[2.2.2]octenyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate 3 O 3 CH H C 3 3 CH N O 3 H N N N O HN N O O 3 O CH O H3C 3 O CH CH CH 3 3 3 x CF3COOH 44.5 mg (0.071 mmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S)- 1-carboxymethoxymethylhexanyl]-N-methyl-L-valinamide mediate 4) were taken up in 10 ml of DMF and admixed successively with 38.6 mg (0.071 mmol) of (2R,3R)methoxy methyl-N-[(2S)(2-oxaazabicyclo[2.2.2]octenyl)oxophenylpropanyl][(2S)- pyrrolidinyl]propanamide trifluoroacetate (Intermediate 197), 32.5 mg (0.086 mmol) of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 41 µl (0.235 mmol) of isopropylethylamine. The mixture was stirred at RT for 1 h. The reaction mixture was then concentrated under reduced pressure and the residue was taken up in ethyl acetate. The organic phase was washed successively with 5% s citric acid solution and 5% aqueous sodium hydrogencarbonate solution, dried over magnesium sulphate, filtered and concentrated. This gave 73 mg (98% of theory) of the Fmoc-protected intermediate N-[(9H- nylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R) methoxymethyl{[(2S)(2-oxaazabicyclo[2.2.2]octenyl)oxophenylpropan yl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide.
HPLC (Method 6): Rt = 2.78 min; LC-MS (Method 3): Rt = 2.96 min; MS (ESIpos): m/z = 1047 (M+H)+. 73 mg (0.071 mmol) of this intermediate were dissolved in 5 ml of DMF. After 0.5 ml of piperidine had been added, the reaction mixture was d at RT for 10 min. This was followed by concentration under reduced pressure, and the residue was digested edly with diethyl ether. After the diethyl ether had been decanted off, the residue was purified by preparative HPLC (eluent: acetonitrile / 0.1% aq. TFA). 16 mg (26% of theory) of the title compound were obtained as a foam.
HPLC (Method 6): Rt = 1.94 min; LC-MS (Method 3): Rt = 1.71 min; MS (ESIpos): m/z = 825 (M+H)+ 1H NMR (400 MHz, DMSO-d6): δ = 8.9-8.6 (m, 3H), 8.4, 8.3, 8.1 and 8.0 (4d, 1H), 7.3-7.1 (m, 5H), 6.7-6.5 (m, 2H), 5.2-4.8 (m, 3H), 4.75-4.55 (m, 3H), 4.05-3.95 (m, 1H), 3.7-3.4 (m, 4H), 3.22, 3.17, 3.15, 3.05, 3.02 and 2.95 (6s, 9H), 3.0 and 2.7 (2 br. m, 2H), 2.46 (m, 3H), 2.4-1.2 (br. m, 13H), 1.1-0.85 (m, 18H), 0.75 (m, 3H) [further s hidden under H2O peak].
Intermediate 199 N-(4-{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S)(2-oxa azabicyclo[2.2.2]octenyl)oxophenylpropanyl]amino}oxopropyl]pyrrolidinyl}- -methyloxoheptanyl]-N-methyl-L-valinamide 3 O O H C 3 CH H C 3 3 CH N O O 3 H H N N N N N N O N N O O O O CH 3 O CH 3 O O CH H C CH CH 3 3 3 3 The title compound was prepared in analogy to Intermediates 193 and 194, proceeding from 23 mg (24 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(2S)(2-oxaazabicyclo[2.2.2]octenyl)oxophenylpropanyl]amino} pyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide oroacetate (Intermediate 198).
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 2): Rt = 2.1 min; MS (ESIpos): m/z = 1118 (M+H)+.
Intermediate 200 N-[2-(2-{2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethoxy)ethyl]-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 N O O H H N O O N N O N O N N O O CH O CH O O H C 3 3 CH 3 O H C CH 3 3 3 The preparation was effected in analogy to Intermediates 174 and 175, commencing with the ive alkylation of Intermediate 192 with Intermediate 172, subsequent deprotection and formation of the maleimide.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 1025 (M+H)+. ediate 201 N-{6-[(bromoacetyl)amino]hexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)- 3-(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl pyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C CH 3 3 3 3 N O H H H N N N N O Br N N O O O CH O CH O O H C 3 3 C CH 3 H CH 3 3 3 22 mg (0.023 mmol) of N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 101) were dissolved in 9.5 ml of THF and admixed at 0°C with 4.2 µl of triethylamine. A solution of bromoacetyl chloride in THF was added dropwise and the on mixture was stirred at 0°C for 30 min. The reaction mixture was concentrated and the residue was purified by preparative HPLC. Thus, 6.9 mg (26% of theory) of the title compound were obtained as a foam.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 11): Rt = 0.9 min; MS (ESIpos): m/z = 1059 and 1061 (M+H)+.
Intermediate 202 2-(2-{3-[(2,5-dioxopyrrolidinyl)oxy]oxopropoxy}ethoxy)ethoxy]ethyl}-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH 3 N O O H H N O O O N N O N O N N O O O CH O CH O O 3 3 CH O H C CH CH 3 3 3 3 The preparation was at first effected in analogy to Intermediate 168, commencing with the reductive tion of Intermediate 192 with ediate 167 and subsequent hydrogenolytic cleavage of the benzyl ester of N-(2-{2-[2-(2-carboxyethoxy)ethoxy]ethoxy}ethyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl tanyl]-N-methyl-L-valinamide. 13 mg (10 µmol) of this intermediate were ved in 5 ml of DMF and admixed with 2.1 mg (20 mmol) of 1-hydroxypyrrolidine-2,5-dione, 6.5 µl of N,N-diisopropylethylamine and 7.1 mg (0.02 mmol) of HATU. The reaction mixture was stirred at RT overnight and then concentrated under reduced pressure. The remaining residue was purified by means of preparative HPLC. After lyophilization from acetonitrile/water, 9.2 mg (62% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 2): Rt = 2.1 min; MS (ESIpos): m/z = 1141 (M+H)+.
Intermediate 203 tert-butyl 6-hydrazinooxohexyl carbamate 3 H O N NH H C N 2 3 H CH O This compound was prepared by rd peptide chemistry methods, by coupling of 6-[(tertbutoxycarbonyl )amino]hexanoic acid with benzyl hydrazinecarboxylate in the presence of EDCI and HOBT, and subsequent hydrogenolytic cleavage of the oxycarbonyl protecting group.
LC-MS (Method 11): Rt = 0.59 min; MS (ESIpos): m/z = 246 (M+H)+.
Intermediate 204 N-{4-[2-(6-aminohexanoyl)hydrazino]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H3C CH H C 3 3 CH 3 NH O O H 2 H H N H N N N N 2 O N N N H O O x CF3COOH O CH3 O CH3 O O CH H3C CH CH 3 3 3 146 mg (50 µmol) of (N-(3-carboxypropyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)- 3-{[(2S)amino(1H-indolyl)oxopropanyl]amino}methoxymethyl pyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were ved in 5 ml of DMF and then admixed with 30.6 mg (80 µmol) of O-(7-azabenzotriazol yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, 19 µl of N,N-diisopropylethylamine and with 22.4 mg (60 µmol) of tert-butyl 6-hydrazinooxohexyl carbamate. The reaction mixture was d at RT for 1.5 h. This was followed by concentration under high vacuum and purification of the remaining residue by means of ative HPLC. Thus, 43 mg (68% of theory) of the protected intermediate were obtained, which were then taken up in 10 ml of dichloromethane and ected with 1 ml of trifluoroacetic acid. The reaction mixture was concentrated and the residue was stirred with dichloromethane, and the solvent was removed again under reduced pressure. Thus, 45 mg (68% of theory over 2 stages) of the title compound were obtained.
HPLC (Method 12): Rt = 1.6 min; LC-MS (Method 11): Rt = 0.66 min; MS (ESIpos): m/z = 983 (M+H)+.
Intermediate 205 N-(4-{2-[6-({[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethyl]carbamoyl}amino)hexanoyl] hydrazino}oxobutyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino (1H-indolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH 3 NH O O O H 2 H H H H N N N N N N O N N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 O 3 3 3 This compound was ed in analogy to Intermediate 114, proceeding from Intermediates 50 and 204.
Yield: 4 mg (78% of ) HPLC (Method 12): Rt = 1.7 min; LC-MS d 11): Rt = 0.73 min; MS (ESIpos): m/z = 1149 (M+H)+.
Intermediate 206 N-(6-{[3-({3-[(2,5-dioxopyrrolidinyl)oxy]oxopropyl}disulphanyl)propanoyl]amino}hexyl)- N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinan- 2-yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 CH3 N O 3 3 O H H H N N S N N N O O S N N O O O O CH3 O CH O O H C C CH CH 3 3 3 3 8 mg (10 µmol) of Intermediate 101 were dissolved in 2 ml of DMF and admixed with 8.6 mg (20 µmol) of 1,1'-{disulphanediylbis[(1-oxopropane-3,1-diyl)oxy]}dipyrrolidine-2,5-dione and 3.7 µl of N,N-diisopropylethylamine. The on mixture was stirred at RT for 2 h and then the solvent was evaporated off under reduced pressure and the residue was purified by preparative HPLC. 7.2 mg (68% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 11): Rt = 0.94 min; MS (ESIpos): m/z = 615 [½ (M+2H+] Intermediate 207 (1S,2R)aminophenylcyclopropanecarboxylic acid trifluoroacetate CF3COOH x O The title compound was obtained in quantitative yield by ecting 210 mg (0.76 mmol) of commercially available (1S,2R)[(tert-butoxycarbonyl)amino]phenylcyclopropanecarboxylic acid with trifluoroacetic acid.
LC-MS (Method 1): Rt = 0.23 min; MS (ESIpos): m/z = 178 .
Intermediate 208 9H-fluorenylmethyl 6-oxohexyl carbamate O NH The title compound was prepared from 1 g (2.95 mmol) of commercially available oren ylmethyl 6-hydroxyhexyl carbamate under standard conditions, by oxidation with sulphur trioxide- pyridine complex. 840 mg (85% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 1.1 min; MS s): m/z = 338 (M+H)+. 2012/075277 Intermediate 209 N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(1S,2R)carboxyphenylcyclopropyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 CH 3 3 OH O 3 H H N N N N O N N O O O CH O CH O O H C 3 3 H C CH CH 3 3 3 3 First, in analogy to the synthesis described in Intermediate 75, by coupling of tbutoxycarbonyl )-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and (1S,2R)aminophenylcyclopropanecarboxylic acid trifluoroacetate (Intermediate 207) in the presence of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, the amine compound N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(1S,2R)carboxyphenylcyclopropyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide was prepared as the trifluoroacetate.
To 22 mg (0.026 mmol) of this compound in 10 ml of methanol were then added 17 mg (0.05 mmol) of 9H-fluorenylmethyl 6-oxohexyl carbamate (Intermediate 208) and 2.3 mg of acetic acid, and also 11.4 mg (0.12 mmol) of borane-pyridine complex. The reaction mixture was stirred at RT overnight. Then the same amounts of borane-pyridine complex and acetic acid, and also 8 mg of fluorenylmethyl 6-oxohexyl carbamate, were added once again and the reaction mixture was stirred at RT for a r 24 h. This was followed by tration under reduced pressure, and the residue was purified by means of preparative HPLC. After concentration of the corresponding fractions, the product was used immediately in the next stage. 33 mg of the still contaminated intermediate were taken up in 5 ml of DMF, and 1 ml of piperidine was added. After stirring at RT for 15 min, the reaction mixture was concentrated and the resulting e was purified by preparative HPLC. Thus, 11 mg (55% of theory over 2 stages) of the aminocarboxylic acid intermediate were ed.
HPLC (Method 12): Rt = 1.7 min; LC-MS (Method 11): Rt = 0.7 min; MS (ESIpos): m/z = 843 . 6 mg (7.12 µmol) of this intermediate were taken up in 1 ml of dioxane and then admixed with 6.6 mg (42.7 µmol) of methyl 2,5-dioxo-2,5-dihydro-1H-pyrrolecarboxylate and with 5 µl of saturated aqueous sodium hydrogencarbonate solution. The reaction mixture was stirred at RT for 1 h. Then another 3 portions each of 50 µl of the saturated aqueous sodium hydrogencarbonate solution were added and the reaction mixture was stirred at RT for a further 30 min. Then the reaction e was acidified to pH 2 with trifluoroacetic acid and subsequently trated under reduced pressure. The ing residue was purified by means of preparative HPLC. After lyophilization from acetonitrile/water, 4 mg (60% of theory) of the title compound were obtained as a foam.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 11): Rt = 0.88 min; MS (ESIpos): m/z = 923 .
Intermediate 210 (2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxy methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide H C CH H C 3 3 3 CH 3 N O O H H N O N N O N N N O O O CH O CH O O CH 3 3 O H C CH CH 3 3 3 3 First, 6-oxohexanoic acid was prepared by a literature method (J. Org. Chem. 58, 1993, 2196- 2200). 80 mg (0.08 mmol) of N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indol- 3-yl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 192) and 65.4 mg (0.5 mmol) of 6-oxohexanoic acid were combined in 9 ml of methanol and admixed with 10 µl of acetic acid and 37.4 mg (0.4 mmol) of borane-pyridine complex. The reaction mixture was stirred at RT overnight. This was followed by concentration under reduced pressure, and the residue was taken up in 1:1 acetonitrile/water and adjusted to pH 2 with trifluoroacetic acid. The reaction mixture was trated again and the residue was purified by means of preparative HPLC. After concentration of the corresponding fractions, 70 mg (86% of theory) of N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S) (1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl pyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were obtained as the trifluoroacetate.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 955 (M+H)+. 1H NMR (500 MHz, DMSO-d 6, characteristic signals): δ = 12.0 (br. M, 1H), 10.8 (s, 1H), 9.4 (m, 1H), 8.9 and 8.8 (2d, 1H), 8.3 and 8.02 (2d, 1H), 7.5 (m, 1H), 7.3 (m, 1H), 7.15 and 7.1 (2s, 1H) 7.05-6.9 (m, 2H), 5.12 and 4.95 (2m, 1H), 4.7-4.5 (m, 2H), 4.1-3.8 (m, 4H), 3.75 (d, 1H), 3.25, 3.2, 3.18, 3.13, 2.98 and 2.88 (6s, 9H), 2.8 (m, 3H), 1.08 and 1.04 (2d, 3H), 0.95-0.8 (m, 15H), 0.8-0.65 (dd, 3H). 22 mg (23 µmol) of this intermediate were dissolved in 1.8 ml of dichloromethane and admixed with 13.2 mg (70 µmol) of 1-(3-dimethylaminopropyl)ethylcarbodiimide hydrochloride, 26.5 mg (230 µmol) of 1-hydroxypyrrolidine-2,5-dione and 0.28 mg (2 µmol) of dimethylaminopyridine, and the on mixture was stirred at RT for 2 h. Subsequently, the reaction mixture was concentrated under reduced pressure and the remaining residue was purified by means of ative HPLC. After lization from itrile/water, 21.3 mg (88% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.94 min; MS (ESIpos): m/z = 1052 (M+H)+.
Intermediate 211 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyl{[(2S,3S)(1,2-oxazinanyl)oxophenylbutan yl]amino}oxopropyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH N O O 3 H H N O N N O N N N O O O CH O CH O O 3 3 CH H C O H C CH CH 3 3 3 3 3 mg (20 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyl{[(2S,3S)(1,2-oxazinanyl)oxophenylbutanyl]amino}oxopropyl] pyrrolidin-yl}methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate (Intermediate 15) were reductively alkylated with exanoic acid, in analogy to Intermediate 210.
Yield: 9.2 mg (61% of theory) HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 929 (M+H)+. 9 mg (10 µmol) of this intermediate were dissolved in 3 ml of DMF and admixed with 5.6 mg (48 µmol) of 1-hydroxypyrrolidine-2,5-dione, 5 µl of N,N-diisopropylethylamine and 5.5 mg (0.015 mmol) of HATU, and the reaction mixture was treated in an ultrasound bath for 6 h. In the course of this, 5.5 mg of HATU were added every hour. Subsequently, the reaction mixture was concentrated under reduced pressure, and the residue was taken up in itrile/water and adjusted to pH 2 with trifluoroacetic acid. After concentrating again under reduced pressure, the remaining e was ed by means of preparative HPLC. After lyophilization from acetonitrile/water, 5.8 mg (57% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.95 min; MS (ESIpos): m/z = 1027 (M+H)+.
Intermediate 212 2-(2-{3-[(2,5-dioxopyrrolidinyl)oxy]oxopropoxy}ethoxy)ethoxy]ethyl}-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S,3S)(1,2- oxazinanyl)oxophenylbutanyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide H3C CH H C 3 3 CH 3 N O O H H N O O O N N O N O N N O O O CH O CH O O 3 3 CH H C H C CH 3 3 O CH 3 3 3 The preparation was at first effected in analogy to Intermediate 168, commencing with the reductive alkylation of Intermediate 15 with Intermediate 167 and subsequent hydrogenolytic cleavage of the benzyl ester of 2-[2-(2-carboxyethoxy)ethoxy]ethoxy}ethyl)-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(2S,3S)(1,2- anyl)oxophenylbutanyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide. 8.4 mg (8 µmol) of this intermediate were dissolved in 3 ml of DMF and admixed with 9.5 mg (80 µmol) of 1-hydroxypyrrolidine-2,5-dione, 10 µl of N,N-diisopropylethylamine and 9.4 mg (25 µmol) of HATU, and the reaction mixture was stirred at RT overnight and then concentrated under reduced pressure. Subsequently, the reaction e was trated under d re, and the residue was taken up in acetonitrile/water and adjusted to pH 2 with trifluoroacetic acid.
After concentrating again under reduced pressure, the remaining residue was purified by means of preparative HPLC. After lyophilization from acetonitrile/water, 4 mg (32% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.96 min; MS (ESIpos): m/z = 1117 (M+H)+.
Intermediate 213 N-{6-[(trans{[(2,5-dioxopyrrolidinyl)oxy]carbonyl}cyclohexyl)amino]oxohexyl}-N- methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinan yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH3 N O H H H N N N N O O N N O O O O CH O CH O O N 3 3 CH H C CH3 CH 3 3 3 O H This compound was prepared in analogy to Intermediate 104, proceeding from N-(5- ypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl) (1,2-oxazinanyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}- 3-methoxymethyloxoheptanyl]-N-methyl-L-valinamide, the sis of which was described under Intermediate 210. 9.3 mg of the title compound (37% of theory over 3 stages) were obtained.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.9 min; MS (ESIpos): m/z = 1177 (M+H)+.
Intermediate 214 N-{4-[(2,5-dioxopyrrolidinyl)oxy]oxobutyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(1S,2R)hydroxyphenylpropanyl]amino}methoxymethyl pyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH O O H H N CH O N N 3 N N N O O O CH O CH O O 3 3 H C OH O H C CH CH 3 3 3 3 This compound was prepared in analogy to Intermediate 210, by conversion of Intermediate 92 to the active ester.
HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 11): Rt = 0.82 min; MS (ESIpos): m/z = 901 (M+H)+.
WO 87716 Intermediate 215 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) R){[(1S,2R)hydroxyphenylpropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH O O 3 H H N CH O N N 3 N N N O O O CH O CH O O 3 3 H C OH H C CH CH 3 O 3 3 3 First, Intermediate 40, in analogy to Intermediate 183, was used with borane-pyridine complex to prepare N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S,2R) hydroxyphenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide. From this compound, in analogy to Intermediate 210, the active ester was then generated. 34 mg (36% of theory over 2 stages) of the title compound were obtained.
HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.85 min; MS (ESIpos): m/z = 930 (M+H)+.
Intermediate 216 N-(4-{[(2,5-dioxopyrrolidinyl)oxy]carbonyl}benzyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)- 2-[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxy- 2-methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide H3C CH H C 3 3 CH N O 3 H N N N O O N N O O O CH3 O CH3 O O CH N H C CH3 CH3 3 O N O First, in analogy to the ation of Intermediate 183, Intermediate 192 was reacted with 4- formylbenzoic acid with borane-pyridine complex to give N-(4-carboxybenzyl)-N-methyl-L-valyl- N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropan- 2-yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan- N-methyl-L-valinamide. This compound was then used, in analogy to Intermediate 210, to generate 11 mg (68% of theory) of the title compound.
HPLC (Method 5): Rt = 1.8 min; LC-MS (Method 1): Rt = 1.13 min; MS (ESIpos): m/z = 1072 (M+H)+.
Intermediate 217 N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)- 1-oxophenylpropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy- -methyloxoheptanyl]-N-methyl-L-valinamide H3C CH H C 3 CH3 O 3 O H H N O N N O N N O O OH CH3 O CH O O CH H CH 3 C CH3 3 3 53 mg (84 µmol) of N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N-[(2R,3S,4S) carboxymethoxymethylhexanyl]-N-methyl-L-valinamide (Intermediate 4) and 45 mg (84 µmol) of benzyl N-{(2R,3R)methoxymethyl[(2S)-pyrrolidinyl]propanoyl}-L- phenylalaninate trifluoroacetate (Intermediate 12) were taken up in 2 ml of DMF, 19 µl of N,N- ropylethylamine, 14 mg (92 µmol) of HOBt and 17.6 mg (92 µmol) of EDC were added and then the mixture was stirred at RT ght. Subsequently, the reaction mixture was concentrated and the residue was purified by means of preparative HPLC. This gave 59 mg (68% of theory) of the Fmoc-protected intermediate N-[(9H-fluorenylmethoxy)carbonyl]-N-methyl-L-valyl-N- S,5S){(2S)[(1R,2R){[(2S)(benzyloxy)oxophenylpropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide.
LC-MS (Method 1): Rt = 1.55 min; m/z = 1044 (M+H)+. 57 mg (0.055 mmol) of this intermediate were treated with 1.2 ml of dine in 5 ml of DMF to detach the Fmoc protecting group. After concentration and purification by means of preparative HPLC, 39 mg (76% of theory) of the free amine intermediate N-methyl-L-valyl-N-[(3R,4S,5S) WO 87716 {(2S)[(1R,2R){[(2S)(benzyloxy)oxophenylpropanyl]amino}methoxymethyl- 3-oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide were ed as the trifluoroacetate.
HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 1.01 min; m/z = 822 (M+H)+. 60 mg (0.06 mmol) of this intermediate were reacted, in analogy to Intermediate 210, with 6- oxohexanoic acid in the presence of borane-pyridine complex. 45 mg (75% of theory) of the title compound were obtained as a foam.
HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.97 min; MS (ESIpos): m/z = 9936 (M+H)+.
Intermediate 218 (2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(benzyloxy)oxophenylpropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 CH 3 O 3 3 O H H N O N N O O N N O O O CH O CH O O CH 3 3 H C CH 3 N CH 3 3 3 O This compound was prepared by conversion of 42 mg (0.05 mmol) of Intermediate 217 to the active ester.
Yield: 26 mg (54%) HPLC d 5): Rt = 2.1 min; LC-MS (Method 1): Rt = 1.01 min; MS (ESIpos): m/z = 1034 (M+H)+.
Intermediate 219 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(1S)carboxyphenylethyl]amino}methoxymethyloxopropyl]pyrrolidin- 1-yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 CH 3 O 3 3 O H H N O N N OH O N N O O O CH O CH O O CH 3 3 N C CH CH 3 3 3 3 mg (0.02 mol) of the nd from Intermediate 218 were taken up in 2.4 ml of methanol and hydrogenated over 5% ium on ted carbon under standard en pressure at RT for min. The st was then filtered off and the solvent was removed under reduced pressure.
The residue was lyophilized from 1:1 acetonitrile/water. This gave 14 mg (92% of theory) of the title compound as a colourless foam.
HPLC (Method 5): Rt = 1.7 min; LC-MS d 1): Rt = 0.86 min; MS (ESIpos): m/z = 944 (M+H)+.
Intermediate 220 N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropan- 2-yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide 3 CH O 3 N H N N 2 O O O CH O O 3 CH H C CH CH 3 3 3 3 500 mg of this intermediate were dissolved in 20 ml of DMF and admixed with 466 mg (3.8 mmol) of Intermediate 191, 382 mg (1.01 mmol) of zabenzotriazolyl)-N,N,N',N'- tetramethyluronium hexafluorophosphate (HATU) and 440 µl (2.5 mmol) of N,N- diisopropylethylamine. The mixture was stirred at RT for 1 h and then concentrated. The residue was taken up in dichloromethane and extracted by shaking first twice with 5% aqueous citric acid solution and then with saturated aqueous sodium hydrogencarbonate solution. The organic phase was concentrated and the residue was purified by flash chromatography on silica gel with 95:5 dichloromethane/methanol as the eluent. The corresponding fractions were combined and the solvent was removed under reduced pressure. After the residue had been dried under high vacuum, 562 mg (65% of theory over both stages) of the Z-protected intermediate were obtained. 562 mg (0.57 mmol) of this intermediate were taken up in 50 ml of methanol and enated with 155 mg of 10% palladium on activated carbon under standard hydrogen pressure at RT for 20 min. The catalyst was then filtered off and the solvent was removed under d pressure. The residue was purified by preparative HPLC. The corresponding fractions were combined, the solvent was evaporated off under d pressure and the residue was lyophilized from dioxane.
This gave 361 mg (87% of theory) of the title nd as a foam.
HPLC (Method 5): double peak with Rt = 1.75 and 1.86 min; LC-MS (Method 1): double peak at Rt = 0.84 min and 0.91 min with the same mass; MS (ESIpos): m/z = 944 .
Intermediate 221 N-{(2S)[(tert-butoxycarbonyl)amino]phenylpropyl}-N-methyl-L-valine H C CH 3 3 3 H O N OH 3 N CH O CH O 3 3 100 mg (0.76 mmol) of commercially available N-methyl-L-valine and 285 mg (1.14 mmol) of commercially available tert-butyl (2S)oxophenylpropanyl carbamate were combined in 22 ml of methanol and admixed with 340 mg (3.66 mmol) of borane-pyridine complex and 70 µl of acetic acid. The reaction mixture was d at RT overnight. This was followed by concentration under reduced pressure, and the residue was ed by flash chromatography on silica gel with dichloromethane/methanol/17% aqueous ammonia solution as the eluent. After concentration of the corresponding ons and lyophilization from 1:1 dioxane/water, 259 mg (93% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 1.6 min; LC-MS (Method 11): Rt = 0.76 min; MS (ESIpos): m/z = 365 (M+H)+.
Intermediate 222 N-[(2S)aminophenylpropyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)- 3-(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C 3 3 3 CH N O 3 H N H N N N O 2 N N O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 x CF3COOH 40 mg (0.11 mmol) of N-{(2S)[(tert-butoxycarbonyl)amino]phenylpropyl}-N-methyl-L- valine (Intermediate 221) were dissolved in 5 ml of DMF and admixed with 80 mg (0.11 mmol) of N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropan- 2-yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide (Intermediate 220), 50 mg (0.13 mmol) of O-(7-azabenzotriazol yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 57 µl (2.5 mmol) of N,N- diisopropylethylamine. The mixture was stirred at RT for 1 h and then concentrated. The residue was taken up in ethyl acetate and washed first with 5% s citric acid solution and then with water. The organic phase was concentrated and the residue was purified by means of preparative HPLC. The corresponding ons were combined and the solvent was removed under reduced pressure. After lyophilization from dioxane, 60 mg (50% of theory) of the protected intermediate were obtained.
HPLC (Method 12): Rt = 2.2 min; LC-MS (Method 1): Rt = 1.17 min; MS (ESIpos): m/z = 1073 (M+H)+. 60 mg (0.05 mmol) of this intermediate were taken up in 10 ml of dichloromethane, 2 ml of trifluoroacetic acid were added, and the reaction mixture was stirred at RT for 1.5 h. Subsequently, the reaction mixture was concentrated under reduced pressure and the remaining residue was ed by means of preparative HPLC. The corresponding fractions were combined, the t was d under reduced pressure and the residue was lyophilized from dioxane/water. In this way, 25 mg (42% of ) of the title compound were obtained as a foam.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.95 min; MS (ESIpos): m/z = 974 (M+H)+.
Intermediate 223 N-[(2S)({[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethyl]carbamoyl}amino)phenylpropyl]- N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinan- 2-yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH O 3 N O H H H H N N N N N O N N N O O O CH O CH O O 3 3 CH O H C CH CH 3 3 3 3 The preparation was effected in analogy to Intermediate 134, proceeding from 5 mg (4.6 µmol) of Intermediate 222. 3.4 mg (65% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 0.99 min; MS s): m/z = 1140 (M+H)+.
Intermediate 224 N-[(2S)({[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethyl]carbamoyl}amino)propyl]-N-methyl- L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH 3 N O O H H H H N N N N N O N N N O O O CH CH O CH O O 3 CH 3 3 O H C CH CH 3 3 3 3 The preparation was effected in y to the sis of Intermediate 223.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.92 min; MS (ESIpos): m/z = 1064 (M+H)+.
Intermediate 225 N-(2-aminoethyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indol yl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidin- 1-yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C 3 3 3 CH 3 N O H H N H N N N 2 O N N O O x CF3COOH CH O CH O O 3 3 CH H C CH CH 3 3 3 3 H 100 mg (0.76 mmol) of commercially ble yl-L-valine and 182 mg (1.14 mmol) of commercially available tert-butyl 2-oxoethyl carbamate were combined in 20 ml of methanol and admixed with 340 mg (3.66 mmol) of borane-pyridine x and 65 µl of acetic acid. The reaction mixture was stirred at RT overnight. This was followed by concentration under reduced pressure, and the residue was purified by flash tography on silica gel with dichloromethane/methanol/17% aqueous ammonia solution (15/4/0.5) as the eluent. After concentration of the corresponding fractions and lyophilization from 1:1 dioxane/water, 190 mg in 39% purity (35% of theory) of the intermediate were obtained, which were converted further without further purification. 50 mg (0.07 mmol) of this intermediate were dissolved in 10 ml of DMF and admixed with 52 mg (0.07 mmol) of N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinan yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 220), 32 mg (0.09 mmol) of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 37 µl (0.2 mmol) of N,N-diisopropylethylamine. The mixture was stirred at RT overnight and then trated. The residue was taken up in ethyl acetate and ted by shaking first with 5% aqueous citric acid solution and then with water. The organic phase was concentrated and the residue was purified by means of preparative HPLC. The corresponding fractions were combined and the solvent was removed under reduced pressure. After lyophilization from dioxane, 53 mg (76% of theory) of the ted intermediate were obtained.
HPLC (Method 12): Rt = 2.0 min; LC-MS d 1): Rt = 1.02 min; MS (ESIpos): m/z = 984 . 53 mg (0.05 mmol) of this intermediate were taken up in 10 ml of dichloromethane, 2 ml of trifluoroacetic acid were added, and the reaction mixture was stirred at RT for 30 min.
WO 87716 Subsequently, the reaction mixture was concentrated under reduced pressure and the remaining residue was purified by means of preparative HPLC. The corresponding fractions were combined, the solvent was removed under reduced pressure and the residue was lyophilized from dioxane/water. In this way, 21 mg (40% of theory) of the title nd were obtained in 65% purity.
HPLC (Method 12): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 884 (M+H)+.
Intermediate 226 N-[2-({[2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethyl]carbamoyl}amino)ethyl]-N-methyl-L-valyl- N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropan- 2-yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan- 4-yl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH 3 N O O H H H H N N N N N O N N N O O O CH O CH O O 3 3 CH O H C CH CH 3 3 3 3 The preparation was effected proceeding from Intermediate 225, in analogy to the synthesis of Intermediate 134. 11.6 mg (59% of theory) of the title compound were obtained.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.90 min; MS s): m/z = 1050 (M+H)+. ediate 227 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(benzyloxy)(1H-indolyl)oxopropanyl]amino}methoxy methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide H C CH H C CH O 3 3 3 3 O H H N O N N O O N N O O O CH O CH O O CH 3 3 3 N H C CH CH 3 3 3 O H This compound was prepared analogously to Intermediate 218, by conversion to the active ester.
Yield: 18 mg (51% of ) HPLC (Method 5): Rt = 2.1 min; LC-MS (Method 1): Rt = 0.98 min; MS (ESIpos): m/z = 1073 .
Intermediate 228 (2R,3S)[(tert-butoxycarbonyl)amino]{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol yl)hexanoyl]hydrazino}oxobutanyl(3R,4S,7S,10S)[(2S)-butanyl]-7,10-diisopropyl(2- {(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl) phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}oxoethyl)-5,11-dimethyl-6,9-dioxo oxa-5,8,11-triazapentadecanoate O CH CH 3 3 O H C CH H C 3 3 3 CH HN O 3 N O O H H H N N N O N N O N N N H O O O O CH O CH O CH O O 3 3 3 CH H C CH3 CH3 3 The title compound was obtained as intermediate in the synthesis of Intermediate 154.
HPLC (Method 12): Rt = 2.1 min; LC-MS (Method 1): Rt = 0.97 min; MS (ESIpos): m/z = 1308 (M+H)+.
Intermediate 229 (2R,3S)acetamido{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino} oxobutanyl (3R,4S,7S,10S)[(2S)-butanyl]-7,10-diisopropyl(2-{(2S)[(1R,2R) methoxymethyl{[(1S,2R)(1,2-oxazinanylcarbonyl)phenylcyclopropyl]amino} oxopropyl]pyrrolidinyl}oxoethyl)-5,11-dimethyl-6,9-dioxooxa-5,8,11-triazapentadecan- CH CH 3 3 O H3C CH H C 3 3 CH N O HN O O 3 H H N N N O N N O N N N H O O O O CH O CH O CH3 O O 3 3 CH H C CH CH 3 3 3 The title compound was prepared from 7.5 mg (2.5 µmol) of Intermediate 154 by acetylation with 2.3 µl of acetic anhydride in 1 ml of DMF in the presence of 0.4 µl of isopropylethylamine.
Yield: 1.4 mg (40% of theory) HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 1250 (M+H)+.
Intermediate 230 (2R,3S)[(tert-butoxycarbonyl)amino]{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl] hydrazino}oxobutanyl (3R,4S,7S,10S)[(2S)-butanyl](2-{(2S)[(1R,2R){[(2S) (1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyloxopropyl] pyrrolidinyl}oxoethyl)-7,10-diisopropyl-5,11-dimethyl-6,9-dioxooxa-5,8,11- triazapentadecanoate O CH CH 3 3 O H C CH H C 3 3 CH N O HN O 3 O 3 H H N N N O N N O N N N H O O O O CH O CH O CH O O 3 3 CH 3 H C CH CH 3 3 3 3 H This nd was prepared in analogy to Intermediate 229, proceeding from Intermediate 193. 16 mg (30% of theory over 3 stages) of the title compound were obtained.
HPLC (Method 12): Rt = 2.0 min; LC-MS (Method 1): Rt = 1.02 min; MS (ESIpos): m/z = 1335 (M+H)+.
WO 87716 Intermediate 231 (2R,3S)acetamido{2-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexanoyl]hydrazino} oxobutanyl (3R,4S,7S,10S)[(2S)-butanyl](2-{(2S)[(1R,2R){[(2S)(1H-indol yl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidin- 1-yl}oxoethyl)-7,10-diisopropyl-5,11-dimethyl-6,9-dioxooxa-5,8,11-triazapentadecan oate CH CH 3 3 O H C CH H C 3 3 3 CH O HN O O 3 N H H N N N O N N O N N N H O O O O CH O CH O CH O O 3 3 3 CH H C CH CH 3 3 3 3 This compound was prepared from 8 mg (6 µmol) of Intermediate 230, first by deprotection with trifluoroacetic acid and subsequent acetylation with acetic anhydride in DMF in the presence of N,N-diisopropylethylamine. 2 mg (37% of theory over 2 stages) of the title compound were obtained.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.88 min; MS (ESIpos): m/z = 1277 (M+H)+. ediate 232 benzyl N-[(4-nitrophenoxy)carbonyl]-beta-alaninate O N O O O 200 mg (0.57 mmol) of commercially available 4-methylbenzenesulphonic enzyl betaalaninate and 229 mg (1.14 mmol) of 4-nitrophenyl chlorocarbonate were taken up in 15 ml of tetrahydrofuran and the reaction mixture was then heated to reflux for 30 min. Subsequently, the reaction mixture was concentrated under reduced pressure and the residue was purified by means of preparative HPLC. After concentration of the corresponding fractions and drying of the residue under high vacuum, 86 mg (44% of theory) of the title compound were obtained.
HPLC d 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 1.07 min; MS (ESIpos): m/z = 345 (M+H)+.
Intermediate 233 N-{2-[({3-[(2,5-dioxopyrrolidinyl)oxy]oxopropyl}carbamoyl)amino]ethyl}-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide H C CH H C O O 3 3 3 CH 3 N N O H H H H N O N N N N O N N O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 13 mg (10 µmol) of ediate 225 and 6.7 mg (20 µmol) of Intermediate 232 were dissolved in 3 ml of DMF, and then 7 µl of N,N-diisopropylethylamine were added. The mixture was stirred at RT overnight and then concentrated under high vacuum. The remaining e was purified by means of preparative HPLC. After concentration of the corresponding fractions and drying of the residue under high vacuum, 5.4 mg (38% of theory) of the protected intermediate were obtained.
HPLC (Method 5): Rt = 2.1 min; LC-MS (Method 1): Rt = 0. 6in; MS (ESIpos): m/z = 1089 (M+H)+. .4 mg (5 µmol) of this intermediate were dissolved in 5 ml of methanol and, after addition of 2 mg of 10% palladium on activated carbon, hydrogenated under standard hydrogen pressure at RT for 20 min. The catalyst was then filtered off and the solvent was removed under reduced pressure.
After the e had been dried under high vacuum, 5 mg (quant.) of the acid intermediate were obtained.
HPLC (Method 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.84 min; MS (ESIpos): m/z = 999 (M+H)+. mg (10 µmol) of this intermediate were ved in 1 ml of DMF and d with 5.8 mg (50 mmol) of 1-hydroxypyrrolidine-2,5-dione and then with 2.6 µl of N,N-diisopropylethylamine and 3.8 mg (10 µmol) of HATU. After ng at RT for 20 h, the reaction mixture was concentrated under reduced pressure. The remaining residue was purified by means of preparative HPLC. After lyophilization from 1:1 dioxane/water, 1.1 mg (20% of ) of the title compound were obtained.
HPLC (Method 12): Rt = 1.9 min; WO 87716 LC-MS (Method 1): Rt = 0.87 min; MS (ESIpos): m/z = 1096 (M+H)+.
Intermediate 234 N-(6-{[(benzyloxy)carbonyl]amino}hexyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 O 3 3 CH O 3 H N H H N O N N N N N N O O O CH O CH O O CH 3 H C CH 3 3 3 CH mg (30 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 55) and 45 mg (180 µmol) of benzyl 6-oxohexyl ate were taken up in 3 ml of methanol and acidified with acetic acid.
At room temperature, 15 µl (144 µmol; 9.4M) of borane-pyridine complex were subsequently added. The mixture was subsequently stirred at RT for 24 h, and acetic acid and 15 µl (144 µmol; 9.4M) of borane-pyridine complex were added again after 8 h. The reaction mixture was uently adjusted to pH 2 with TFA and purified by means of preparative HPLC. The product fractions were combined and trated, and the residue was dried under high vacuum. This gave 15 mg (46% of theory) of the title compound as a foam.
LC-MS (Method 1): Rt = 1.03 min; m/z = 1066 (M+H)+.
Intermediate 235 N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 CH O O 3 H N H N H N N N N 2 N N O O CH O CH O O CH 3 3 3 H C CH 3 3 CH mg (14 µmol) of [(benzyloxy)carbonyl]amino}hexyl)-N-methyl-L-valyl-N-[(3R,4S,5S)- 3-methoxy{(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl(5-phenyl-1,3,4- oxadiazolyl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide (Intermediate 234) were taken up in 3 ml of methanol, and 1.8 mg of palladium on al (5%) were added. The reaction mixture was subsequently hydrogenated under standard hydrogen pressure at RT for 2 h. The catalyst was then filtered off and the solvent was removed under d pressure. The residue was lyophilized from 1:1 acetonitrile/water. 11 mg (86% of ) of the title compound were obtained as a foam.
LC-MS (Method 1): Rt = 0.81 min; m/z = 932 (M+H)+.
Intermediate 236 N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 O 3 3 CH O 3 H N H N N N N N N N O O O CH O CH 3 O O CH 3 H C CH 3 3 CH 11 mg (12 µmol) of N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S) [(1R,2R)methoxymethyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 235) were taken up in 500 µl of 1:1 dioxane/water and admixed with 253 µl of 1M aqueous sodium hydrogencarbonate solution and then with 2.8 mg (18 µmol) of methyl 2,5-dioxo- 2,5-dihydro-1H-pyrrolecarboxylate. The reaction mixture was stirred at RT for 30 min and then acidified with trifluoroacetic acid. The reaction mixture was purified by means of preparative HPLC. After lyophilization, 0.8 mg (7% of theory) of the title compound was obtained.
LC-MS d 1): Rt = 1.01 min; m/z = 1012 (M+H)+.
Intermediate 237 N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy- 2-methyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 O 3 3 CH O 3 H N H N HO N N N N N O O O CH O CH O CH 3 H C CH 3 O 3 3 3 CH mg (30 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxy methyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidin- 1-yl}methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 55) and 23 mg (180 µmol) of exanoic acid were taken up in 3 ml of methanol and acidified with acetic acid. At room temperature, 15 µl (144 µmol; 9.4M) of borane-pyridine complex were subsequently added. The reaction mixture was subsequently stirred at RT for 20 h, and acetic acid and 15 µl (144 µmol; 9.4M) of borane-pyridine complex were added again after 8 h. The reaction mixture was subsequently adjusted to pH 2 with trifluoroacetic acid and ed by means of preparative HPLC. The product fractions were ed and trated, and the residue was lyophilized. 21 mg (74% of theory) of the title compound were thus obtained as a foam.
LC-MS (Method 1): Rt = 0.91 min; m/z = 947 (M+H)+.
Intermediate 238 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L- valinamide O O H C CH 3 O 3 3 CH N O 3 H N H N O N N N N N O O O CH O CH O CH 3 H 3 O 3 C CH 3 3 CH 21 mg (22 µmol) of Intermediate 237 were dissolved in 1 ml of DMF and admixed with 38 mg (333 µmol) of 1-hydroxypyrrolidine-2,5-dione and then with 2.4 mg (10 µmol) of O-(7- zotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 19 µl of N,N-diisopropylethylamine. After stirring at RT for 2 h, the reaction mixture was purified by means of preparative HPLC. After lyophilization from dioxane, 22 mg (96% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.95 min; m/z = 1044 (M+H)+.
Intermediate 239 N-methyl-L-threonyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2- oxazinanyl)oxopropanyl]amino}methoxymethyloxopropyl] pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H3C OH H C 3 CH 3 N O H H N N N O HN N O O CH O CH O O 3 3 CH3 H C CH CH3 3 3 First, N-[(benzyloxy)carbonyl]-N-methyl-L-threonine was released from 237 mg (0.887 mmol) of its dicyclohexylamine salt by taking it up in ethyl acetate and extractive g with 5% aqueous sulphuric acid. The organic phase was dried over magnesium sulphate, filtered and concentrated. 14.7 mg (0.055 mmol) of N-[(benzyloxy)carbonyl]-N-methyl-L-threonine were taken up in 3 ml of DMF and d successively with 40 mg (0.055 mmol) of Intermediate 220, 12.7 mg (0.066 mmol) of imethylaminopropyl)ethylcarbodiimide hydrochloride and 10 mg (0.066 mmol) of 1-hydroxy-1H-benzotriazole e. The mixture was subsequently stirred at RT for 2 h. The solvent was then removed under reduced pressure and the residue purified by preparative HPLC. 29 mg (54% of theory) of the Z-protected intermediate were thus obtained.
LC-MS (Method 1): Rt = 1.15 min; MS (ESIpos): m/z = 976 (M+H)+. 29 mg (0.003 mmol) of this intermediate were ved in 5 ml of methanol and hydrogenated over 5 mg of 5% palladium/charcoal at RT and standard re for 1 h. The catalyst was subsequently filtered off and the solvent was evaporated off. The remaining residue was purified by preparative HPLC. 17 mg (54% of theory) of the title compound were obtained.
LC-MS (Method 1): Rt = 0.77 min; MS (ESIpos): m/z = 842 (M+H)+.
Intermediate 240 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-threonyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxy methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C OH H C 3 3 CH 3 N O O H H N O N N O N N N O O O CH O CH O O 3 3 CH O H C CH CH 3 3 3 3 This compound was prepared in analogy to Intermediate 210 from 15.6 mg (0.016 mmol) of Intermediate 239. 10.8 mg (67% of theory over 2 stages) of the title compound were ed.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.85 min; MS (ESIpos): m/z = 1053 .
Intermediate 241 N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(4-hydroxyphenyl)(1,2-oxazinan- 1-oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH H C CH N 3 3 3 O 3 H H N N N O HN N O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 First, in analogy to Intermediate 5, trifluoroacetic acid-(2S)amino(4-hydroxyphenyl)(1,2- oxazinanyl)propanone (1:1) was prepared. This reagent was then used, in analogy to the synthesis described in Intermediate 75, by ng with N-(tert-butoxycarbonyl)-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxy- -methyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) in the presence of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium uorophosphate and subsequent detachment of the Boc protecting group by means of trifluoroacetic acid, to e the title compound.
HPLC (Method 12): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.85 min; MS (ESIpos): m/z = 817 (M+H)+.
Intermediate 242 N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(4-hydroxyphenyl)(1,2-oxazinanyl)oxopropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- namide H C CH H C 3 3 3 CH O 3 N O H H N O N N O N N N O O O CH O CH O O 3 3 CH O H C CH CH 3 3 50 mg (0.05 mmol) of Intermediate 241 were reacted, in analogy to Intermediate 210, with 6- oxohexanoic acid in the presence of borane-pyridine complex. Subsequently, 22.5 mg (0.02 mmol) of the resulting acid were converted to the activated ester. 13.5 mg (36% of theory over 2 stages) of the title compound were obtained.
HPLC (Method 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 1028 (M+H)+.
Alternatively, the title compound can also be obtained by one-hour catalytic hydrogenation of Intermediate 250 at room temperature in ol over 10% palladium on active carbon under hydrogen standard pressure.
Intermediate 243 N-(6-aminohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(4- hydroxyphenyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH N O 3 H N H N N N 2 O N N O O CH O CH O O H C 3 3 H C CH 3 3 3 3 OH The preparation was effected in analogy to ediate 78, by reductive alkylation of Intermediate 241 with benzyl exyl carbamate and borane-pyridine complex and subsequent hydrogenation in methanol as the solvent.
Yield: 17.5 mg (34% of theory over 2 stages) HPLC (Method 12): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.63 min; MS (ESIpos): m/z = 916 .
Intermediate 244 N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)hexyl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(4-hydroxyphenyl)(1,2-oxazinanyl)oxopropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide O H3C CH H C CH3 3 3 N O H H N N N N O N N O O O CH O CH O O H C 3 3 3 H3C CH CH 3 3 The preparation was effected in analogy to Intermediate 166, proceeding from Intermediate 243.
Yield: 1.3 mg (12% of theory) HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.89 min; MS (ESIpos): m/z = 996 (M+H)+. ediate 245 2,5-dioxopyrrolidinyl ,4S,7S,10S)[(2S)-butanyl](2-{(2S)[(1R,2R){[(2S) (1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}oxoethyl)-7,10-diisopropyl-5,11-dimethyl-6,9,15-trioxooxa- 5,8,11-triazapentadecanyl]-N-(tert-butoxycarbonyl)-L-threonyl-beta-alaninate O CH CH 3 3 H C CH H C 3 3 3 CH HN O 3 N O O H H H N O N O N N O N N N O O O O CH O CH O CH O O 3 3 3 CH H C CH CH 3 O 3 3 3 First, Intermediate 193, as described under Intermediate 154, was reacted with benzyl N-(tertbutoxycarbonyl )-L-threoninate and then the benzyl ester was removed by hydrogenolysis. 30 mg (0.027 mmol) of the N-[4-({(1S,2R)[(tert-butoxycarbonyl)amino]carboxypropanyl}oxy) yl]-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2- oxazinanyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide thus obtained were then coupled with 4-methylbenzenesulphonic acid-benzyl beta-alaninate (1:1) in the ce of HATU and the benzyl ester was removed again by hydrogenolysis (yield: 24 mg (71% of theory over 2 stages)). Finally, 10 mg (0.008 mmol) of the resulting acid were converted to the activated ester. After HPLC purification, 2.7 mg (23% of theory) of the title compound were obtained.
HPLC (Method 5): Rt = 1.9 min; LC-MS (Method 1): Rt = 1.01 min; MS (ESIpos): m/z = 1295 (M+H)+ Intermediate 246a (2S)amino(4-hydroxy-1,2-oxazolidinyl)(1H-indolyl)propanone trifluoroacetic acid (1:1) Diastereomer 1 CF3COOH x H N 2 O Diastereomer 1 CF3COOH x H N 2 O Diastereomer 2 1.6 g (3.982 mmol) of 2,5-dioxopyrrolidinyl N-(tert-butoxycarbonyl)-L-tryptophanate were dissolved in 15 ml of DMF and admixed with 500 mg (3.982 mmol) of 1,2-oxazolidinol and 100 µl of N,N-diisopropylethylamine. The reaction mixture was stirred at RT ght. Then another 100 µl of N,N-diisopropylethylamine were added, and the mixture was first d in an ultrasound bath for 5 h, then stirred at RT overnight, and subsequently concentrated under reduced pressure. The remaining residue was taken up in ethyl acetate and shaken first twice with 5% citric acid solution, then with ted sodium hydrogencarbonate solution and finally with water. The organic phase was trated and the e was separated into the reomers by flash chromatography on silica gel with 95:5 dichloromethane/methanol as the eluent. The corresponding fractions of both diastereomers were combined and the solvent was removed under reduced pressure. After the residues had been dried under high vacuum, 272 mg (18% of theory) of Diastereomer 1 (Rf = 0.18 (95:5 dichloromethane/methanol) and 236 mg (16% of theory) of Diastereomer 2 (Rf = 0.13 (95:5 dichloromethane/methanol), and also 333 mg (22% of theory) of a mixed fraction of the Boc-protected intermediates were obtained.
Under standard conditions, 5 ml of trifluoroacetic acid in 20 ml of dichloromethane were used to detach the Boc ting group from 272 mg (725 µmol) of Diastereomer 1 of this intermediate and, after lyophilization from dioxane/water, 290 mg (quant) of the title compound were ed in 75% purity and used in the next stage without further purification.
HPLC (Method 12): Rt = 1.1 min; LC-MS d 13): Rt = 1.80 min; MS (ESIpos): m/z = 276 (M+H)+ Intermediate 246b (2S)amino(4-hydroxy-1,2-oxazolidinyl)(1H-indolyl)propanone trifluoroacetic acid (1:1) Diastereomer 2 CF3COOH x H N 2 O Diastereomer 2 Under standard conditions, 5 ml of trifluoroacetic acid in 20 ml of dichloromethane were used to detach the Boc protecting group from 236 mg (630 µmol) of Diastereomer 2 of the intermediate described in 246a and, after tration, stirring with diethyl ether and drying of the residue under high vacuum, 214 mg (76%) of the title nd were obtained.
LC-MS (Method 13): Rt = 1.84 min; MS (ESIpos): m/z = 276 (M+H)+ Intermediate 247a N-{6-[(2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(4-hydroxy-1,2-oxazolidinyl)(1H-indolyl)oxopropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide 3 O H C CH H C 3 3 3 CH N O O 3 H H N O N N O N N N O O O CH O CH O O 3 3 CH O H C CH CH 3 3 3 3 Diastereomer 1 To synthesize this compound, the coupling of Intermediates 26 and 246a with subsequent detachment of the Boc protecting group was first med as described for Intermediate 74.
Subsequently, the alkylation with 6-oxohexanoic acid in the presence of borane-pyridine x and subsequent conversion of the acid to the active ester were performed as described in Intermediate 210. The title nd was purified by preparative HPLC.
HPLC (Method 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 1053 (M+H)+ Intermediate 247b (2,5-dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)(4-hydroxy-1,2-oxazolidinyl)(1H-indolyl)oxopropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide 3 O H C CH H C 3 3 3 CH 3 N O O H H N O N N O N N N O O O CH O CH O O 3 3 CH C CH CH 3 O H 3 3 3 Diastereomer 2 To synthesize this compound, the coupling of Intermediates 26 and 246b with subsequent detachment of the Boc protecting group was first performed as described for Intermediate 74.
Subsequently, the tion with exanoic acid in the presence of borane-pyridine complex and subsequent conversion of the acid to the active ester were performed as described in Intermediate 210. The title compound was purified by preparative HPLC.
HPLC d 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 1053 (M+H)+ Intermediate 248 N-(5-carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)tert-butoxy- ydroxyphenyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}- 3-methoxymethyloxoheptanyl]-N-methyl-L-valinamide CH3 3 H C CH H3C CH CH 3 O 3 O 3 3 H N HO N N O N N O O O CH O CH O O H C 3 3 CH 3 H C CH 3 3 3 First, in analogy to the synthesis described in Intermediate 86, by coupling N-(tert- butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R)carboxy methoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide (Intermediate 26) and utyl L-tyrosinate in the presence of zabenzotriazolyl)- N,N,N',N'-tetramethyluronium hexafluorophosphate and subsequent detachment of the Boc protecting group by means of oroacetic acid to obtain the tert-butyl ester (stirring with oroacetic acid in dichloromethane for 40 min), the amine compound tert-butyl N-[(2R,3R) methoxy{(2S)[(3R,4S,5S)methoxymethyl(methyl{(2S)methyl[(N-methyl-L- valyl)amino]butyl}amino)heptanoyl] pyrrolidinyl}methylpropanoyl]-L-tyrosinate was prepared as the trifluoroacetate. 38 mg (0.04 mmol) of this compound were then used, in analogy to the preparation of Intermediate 210, by reaction with 4-oxohexanoic acid in the presence of borane- pyridine complex, to obtain 31 mg (99% of theory) of the title compound.
HPLC (Method 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.88 min; MS (ESIpos): m/z = 918 (M+H)+.
Intermediate 249 Trifluoroacetic acid-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)[4-(benzyl- oxy)phenyl](1,2-oxazinanyl)oxopropanyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide(1:1) OH CH3 F H C CH H C CH N 3 3 3 O 3 H N N N O HN N O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 First, in analogy to ediates 5 and 6, starting from O-benzyl-N-(tert-butoxycarbonyl)-L- tyrosine, trifluoroacetic acid-(2S)amino[4-(benzyloxy)phenyl](1,2-oxazinanyl)propan- 1-one(1:1) was prepared. Then, from this building block, in analogy to the synthesis described in Intermediate 75, coupling with N-(tert-butoxycarbonyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)- 2-[(1R,2R)carboxymethoxypropyl]pyrrolidinyl}methoxymethyloxoheptanyl]- N-methyl-L-valinamide (Intermediate 26) in the presence of O-(7-azabenzotriazolyl)-N,N,N',N'- tetramethyluronium hexafluorophosphate and subsequent elimination of the Boc protecting group by means of trifluoroacetic acid gave the title compound.
HPLC (Method 12): Rt = 2.15 min; LC-MS (Method 1): Rt = 0. 99 min; MS (ESIpos): m/z = 908 (M+H)+.
Intermediate 250 N-{6-[(2,5-Dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) [(1R,2R){[(2S)[4-(benzyloxy)phenyl](1,2-oxazinanyl)oxopropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide H3C CH H3C 3 CH 3 N O O H H N O N N O N N N O O O CH O CH3 O O 3 CH3 H3C CH3 CH3 100 mg (0.088 mmol) of ediate 249 were d in analogy to Intermediate 210 with 6- oxanoic acid in the presence of borane-pyridine complex. Then 30 mg (0.029 mmol) of the 2012/075277 resulting acid were converted into the activated ester. This gave 15 mg (40% of theory over 2 stages) of the title compound.
HPLC (Method 12): Rt = 2.26 min; LC-MS (Method 1): Rt = 1.05 min; MS (ESIpos): m/z = 1119 (M+H)+.
Intermediate 251 N-[4-(2-{5-[(2,5-Dioxopyrrolidinyl)oxy]oxopentanoyl}methylhydrazino)oxobutyl]-N- methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinan yl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide O H C CH H C 3 CH N O 3 3 O O 3 H H N N N N N O O N N N O O O CH O CH O CH O O 3 3 3 CH H C CH CH 3 3 3 3 H First of all, 30 mg (0.032 mmol) of Intermediate 193 were converted into the activated N- hydroxysuccinimide ester. 10.3 mg (0.009 mmol) of this active ester were dissolved in 2 ml of DMF, admixed with 2.7 mg (0.018 mmol) of utyl 1-methylhydrazinecarboxylate and 8 µL N,N-diisopropylethylamine, and stirred at RT for 16 h. This operation was repeated, and then the batch was concentrated and the residue which remained was purified by preparative HPLC.
Concentration of the corresponding fractions and drying under a high vacuum gave 5.4 mg (43%) of the intermediate.
LC-MS (Method 1): Rt = 0. 99 min; MS (ESIpos): m/z = 1054 (M+H)+.
The Boc protecting group was removed from 3.5 mg (0.002 mmol) of this intermediate, using trifluoroacetic acid in dichloromethane. Following concentration and drying under a high vacuum, the residue was taken up in 4 ml of romethane and admixed with 1,1'-[(1,5-dioxopentane- 1,5-diyl)bis(oxy)]dipyrrolidine-2,5-dione and 2 µl of N,N-diisopropylethylamine. After ng at RT for 1 h, the batch was trated and the residue which remained was ed by preparative HPLC. Concentration of the corresponding fractions and drying under a high vacuum gave 1.4 mg (44%) of the title compound.
HPLC (Method 5): Rt = 1.6 min; LC-MS d 1): Rt = 0.88 min; MS (ESIpos): m/z = 1166 (M+H)+.
Intermediate 252 N-(2-{2-[2-(2-Carboxyethoxy)ethoxy]ethoxy}ethyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) R){[(2S)(4-hydroxyphenyl)(1,2-oxazinanyl)oxopropanyl]amino}methoxy- 2-methyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide H C CH H C CH 3 3 3 3 N O H H N HO O O N N O O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 100 mg (0.088 mmol) of Intermediate 249 and 109 mg (0.350 mmol) of Intermediate 167 were combined in 10 ml of ol and admixed with 39 mg (0.42 mmol) of borane-pyridine complex and 15 µl of acetic acid. The batch was stirred at RT overnight. Then the same amounts of boranepyridine complex and of acetic acid were added once again, and the batch was stirred for a further 24 h at RT. It was then concentrated under reduced pressure, and the residue was purified by means of preparative HPLC. Concentration of the ponding fractions and lyophilization from dioxane/water 1:1 gave 98 mg (93% of theory) of the bis-benzyl ediate. This intermediate was taken up in 18.5 ml of methanol and subjected to catalytic hydrogenation over 5% palladium on activated carbon under standard hydrogen pressure at room temperature for 1 h. Filtration, concentration, and lyophilization of the residue from dioxane gave 73 mg (87% of theory) of the title compound.
HPLC (Method 12): Rt = 1.85 min; LC-MS (Method 1): Rt = 0.84 min; MS (ESIpos): m/z = 1021 .
Intermediate 253 N-{2-[2-(2-{3-[(2,5-Dioxopyrrolidinyl)oxy]oxopropoxy}ethoxy)ethoxy]ethyl}-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(4-hydroxyphenyl)(1,2-oxazinanyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide 2012/075277 O O H C CH CH3 N N 3 3 3 O H H N O O O N N O O N N O O O CH3 O CH O O 3 CH3 H C CH CH3 3 3 22 mg (22 µmol) of Intermediate 252 were dissolved in 8.5 ml of DMF and admixed with 25 mg (215 µmol) of 1-hydroxypyrrolidine-2,5-dione and subsequently with 12.3 mg (32 µmol) of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 37 µl of N,N-diisopropylethylamine. After ng at RT for 2 h, the reaction e was purified by means of preparative HPLC. Lyophilization from dioxane gave 16 mg (62% of theory) of the title compound.
HPLC (Method 5): Rt = 1.57 min; LC-MS d 1): Rt = 0.8 min; MS (ESIpos): m/z = 1118 (M+H)+.
Intermediate 254 N-(2-{2-[2-(2-Carboxyethoxy)ethoxy]ethoxy}ethyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide O N HO O O N N N O N N O O O O O O 10 mg (0.012 mmol) of Intermediate 55 and 11 mg (0.036 mmol) of Intermediate 167 were combined in 1 ml of methanol and admixed with 5.4 mg (0.058 mmol) of borane-pyridine complex and 1 µl of acetic acid. The batch was stirred at RT overnight. Then the same amounts of boranepyridine complex and of acetic acid were added once again, and the batch was stirred at RT for a further 20 h. It was then concentrated under d pressure and the residue was purified by means of preparative HPLC. Concentration of the corresponding fractions and lyophilization from dioxane/water 1:1 gave 8 mg (58% of theory) of bis-benzyl intermediate. This intermediate was taken up in 2 ml of methanol and subjected to catalytic hydrogenation over 5% palladium on activated carbon under standard en pressure at room temperature for 1 h. Filtration, concentration, and lyophilization of the residue from dioxane gave 7 mg (95% of theory) of the title compound.
LC-MS d 1): Rt = 0.99 min; MS (ESIpos): m/z = 1036 (M+H)+.
Intermediate 255 N-{2-[2-(2-{3-[(2,5-Dioxopyrrolidinyl)oxy]oxopropoxy}ethoxy)ethoxy]ethyl}-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl (5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N- methyl-L-valinamide O N O O O N O O O N N N O N N O O O O O O 7.3 mg (7 µmol) of Intermediate 254 were dissolved in 0.3 ml of DMF and admixed with 12 mg (106 µmol) of 1-hydroxypyrrolidine-2,5-dione and subsequently with 13.5 mg (35 µmol) of O-(7- azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 6 µl of N,N-diisopropylethylamine. After ng at RT for 2 h, the reaction mixture was purified by means of preparative HPLC. lization from dioxane gave 7.7 mg (79% of theory) of the title compound.
LC-MS (Method 1): Rt = 0.97 min; MS (ESIpos): m/z = 1134 (M+H)+. ediate 256 N-(2-{2-[2-(2-Aminoethoxy)ethoxy]ethoxy}ethyl)-N-methyl-L-valyl-N-[(3R,4S,5S)methoxy {(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl(5-phenyl-1,3,4-oxadiazol yl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N-methyl-L-valinamide O N O O N N N H N N N 2 O O O O O O mg (0.02 mmol) of ediate 55 were subjected to reductive amination in analogy to the preparation of Intermediate 254, with benzyl (2-{2-[2-(2-oxoethoxy)ethoxy]ethoxy}ethyl)- carbamate (Intermediate 172) in the presence of borane-pyridine complex. The Z protecting group was subsequently removed by hydrogenolysis, using 5% palladium on carbon as catalyst and in methanol as solvent, and 21 mg (85% of theory over 2 stages) of the title compound were prepared.
LC-MS (Method 1): Rt = 0.85 min; MS (ESIpos): m/z = 1008 (M+H)+.
Intermediate 257 2-{2-[2-(2,5-Dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy]ethoxy}ethoxy)ethyl]-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyloxo{[(1S)phenyl (5-phenyl-1,3,4-oxadiazolyl)ethyl]amino}propyl]pyrrolidinyl}methyloxoheptanyl]-N- methyl-L-valinamide O O N O O N N N N O N N O O O O O 21 mg (20.8 µmol) of ediate 256 were taken up in 1 ml of dioxane/water 1:1 and then admixed with 4.9 mg (31.2 µmol) of methyl 2,5-dioxo-2,5-dihydro-1H-pyrrolecarboxylate and also wtih 42 µl of 1M s sodium hydrogen carbonate solution. The on mixture was stirred at RT for 30 min. Then a further 374 µl of the 1M aqueous sodium hydrogen carbonate solution were added, and the reaction mixture was d at RT for a r 30 min and then concentrated under reduced pressure. The residue which remained was purified by means of preparative HPLC. Lyophilization gave 4.5 mg (20% of theory) of the title compound, as a colourless foam.
LC-MS (Method 1): Rt = 1.04 min; MS (ESIpos): m/z = 1088 (M+H)+.
Intermediate 258 N-{2-[2-(2-{3-[(2,5-Dioxopyrrolidinyl)oxy]oxopropoxy}ethoxy)ethoxy]ethyl}-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)[4-(benzyloxy)phenyl](1,2-oxazinanyl)- 1-oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl- 1-oxoheptanyl]-N-methyl-L-valinamide O O H C CH H C 3 3 CH 3 O 3 N N H H N O O O N N O O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 This compound was prepared starting from Intermediate 249 by reductive alkylation using tertbutyl 3-{2-[2-(2-oxoethoxy)ethoxy]ethoxy}propanoate, followed by t-butyl ester ge and conversion into the N-hydroxysuccinimide ester.
HPLC (Method 5): Rt = 1.96 min; LC-MS (Method 1): Rt = 1.11 min; MS (ESIpos): m/z = 1208 (M+H)+.
Intermediate 259 N-(5-Carboxypentyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3E)-1,4- diphenylbutenyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy methyloxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH O 3 H H N HO N N N N O O O CH O CH3 O O CH 3 H C CH3 3 CH 9.6 mg (8.4 µmol) of N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S,3E)-1,4- diphenylbutenyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxy oxoheptanyl]-N-methyl-L-valinamide trifluoroacetate (Intermediate 58) and 6.5 mg (51 µmol) of 6-oxohexanoic acid were taken up in 769 µl of methanol and acidified with acetic acid. Then, at room temperature, 4 µl (40 µmol) of borane-pyridine x were added. The reaction mixture was subsequently stirred at RT for 20 h. The reaction mixture was then adjusted to a pH of 2 with trifluoroacetic acid, and purified by means of preparative HPLC. The production fractions were combined and trated and the residue was lized. This gave 8 mg (93% of theory) of the title compound, as a foam.
LC-MS (Method 1): Rt = 1.06 min; m/z = 1019 (M+H)+.
WO 87716 Intermediate 260 N-{6-[(2,5-Dioxopyrrolidinyl)oxy]oxohexyl}-N-methyl-L-valyl-N-[(3R,4S,5S){(2S) R){[(2S,3E)-1,4-diphenylbutenyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide O O H C CH H C N 3 3 3 CH O 3 H H N O N N N N O O O CH O CH O O CH 3 H C CH 3 3 3 CH 7.5 mg (7.4 µmol) of Intermediate 259 were dissolved in 332 µl of DMF and admixed with 12.7 mg (110 µmol) of 1-hydroxypyrrolidine-2,5-dione and subsequently with 14 mg (37 µmol) of O-(7-azabenzotriazolyl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 6 µl of N,N-diisopropylethylamine. After stirring at RT for 2 h, the reaction mixture was purified by means of preparative HPLC. lization from dioxane gave 4 mg (55% of theory) of the title compound.
LC-MS (Method 1): Rt = 1.19 min; m/z = 1002 (M+H)+.
Intermediate 261 N-{2-[2-(2-{3-[(2,5-Dioxopyrrolidinyl)oxy]oxopropoxy}ethoxy)ethoxy]ethyl}-N-methyl-L- valyl-N-[(3R,4S,5S)methoxy{(2S)[(1R,2R)methoxymethyl{[(1S,2R)(1,2- oxazinanylcarbonyl)phenylcyclopropyl]amino}oxopropyl]pyrrolidinyl}methyl oxoheptanyl]-N-methyl-L-valinamide H C CH H C 3 3 3 CH 3 N O O H H N O O O N N O N O N N O O O CH O CH O O 3 3 CH CH CH 3 O H C 3 3 3 This compound was prepared starting from Intermediate 16 by reductive tion with benzyl 3- {2-[2-(2-oxoethoxy)ethoxy]ethoxy}propanoate, subsequent benzyl ester cleavage by hydrogenolysis, and conversion into the N-hydroxysuccinimide ester.
HPLC (Method 5): Rt = 1.83 min; LC-MS (Method 1): Rt = 0.93 min; MS s): m/z = 1114 (M+H)+.
B: Preparation of antibody-drug conjugates (ADCs) B-1. Generation of anti-FGFR2 antibodies The human anti-FGFR2 antibodies M048-D01-hIgG1 and M048-D01-hIgG1-b of the present invention were isolated by phage display logy, using the naïve Fab antibody library n-CoDeR from Bioinvent International AB (Lund, Sweden; described in Söderling et al., Nat.
Biotech. 2000, 18:853-856). One ing hit was the parental Fab fragment M048-D01. The variable region of the heavy chain Vh of this Fab fragment is given by SEQ ID NO: 21; the variable region of the light chain Vl is given by SEQ ID NO: 22. Following identification of the 01 Fab fragment, it was expressed under the name M048-D01-hIgG1-b (SEQ ID NO: 10 for the heavy chain, and SEQ ID NO: 9 for the light chain) in the form of human IgG. For efficient g, the first three amino acids of the N-terminus of the heavy chain of M048-D01-hIgG1-b [EVQ] were also expressed, alternatively, as QVE. This variation gave the antibody M048-D01- hIgG1 (SEQ ID NO: 8 for the heavy chain and SEQ ID NO: 7 for the light chain). The variable regions Vh and Vl of M048-D01-hIgG1 are given by SEQ ID NO: 11 and SEQ ID NO: 12; the variable regions Vh and Vl of M048-D01-hIgG1-b are given by SEQ ID NO: 13 and SEQ ID NO: 14. Both antibodies have the same CDR sequences, which are given by SEQ ID NOs: 15 (H-CDR1), 16 (H-CDR2), 17 (H-CDR3), 18 (L-CDR1), 19 2) and 20 (L-CDR3).
The antibodies 21-mIgG1 and GAL-FR22-mIgG2a are described in WO2010/054265 as mIgG1 (GAL-FR21) and mIgG2b (GAL-FR22). GAL-FR21 is defined therein via Vh (SEQ ID NO: 4 in WO2010/054265) and via Vl (SEQ ID NO: 1 in WO2010/054265), and GAL-22 is defined via Vh (SEQ ID NO: 8 in WO2010/054265) and via Vl (SEQ ID NO: 7 in WO2010/054265). For the studies bed in the present application, Vl and Vh of GAL-FR21 were reformatted into the murine IgG1 format, resulting in the antibody GAL-FR21-mIgG1 (SEQ ID NO: 3 and SEQ ID NO: 4 of the present ation). Vl and Vh of GAL-FR22 were reformatted into the murine IgG2a format, resulting in the antibody GAL-FR22-mIgG2a (SEQ ID NO: 5 and SEQ ID NO: 6 of the present application).
B-2. Expression of anti-FGFR2 antibodies The dies M048-D01-hIgG1, M048-D01-hIgG1-b, GAL-FR21-mIgG1 and GAL-FR22- mIgG2a were produced transiently in mammalian cell culture.
The expression constructs were produced as described below: In order to transfer the Vh and Vl regions of the M048-D01 Fab originating from the phage display into the IgG , and in order to change the sion system from E. coli to mammalian cells, the Vh and Vl sequences from the phage display E. coli clone were amplified using PCR primers.
The ng restriction enzyme cleavage sites were uced at the 5’ and 3’ ends both of Vh and of Vl. These restriction enzyme cleavage sites were used for the cloning of Vh and Vl into an expression vector which contained the IgG backbone.
The E. coli cells were incubated in a test tube with 100 µl of water at 95°C for 10 minutes and then placed on ice for 5 minutes. Following brief vortexing, the solution was clarified by centrifugation.
The supernatant separated off was used for DNA amplification. The PCR reactions were carried out tely for Vh and Vl. This was done using specific primer pairs with BamHI and NotI cleavage sites for Vl and MfeI, and MfeI and BlpI cleavage sites for Vh. The PCR reactions were carried out using the ime Pfx rase (Invitrogen, # 12344-024) according to manufacturer instructions. The PCR products were analysed on 1% agarose gels. To obtain compatible ends, the expression vectors and PCR products were digested according to cturer instructions with the corresponding restriction endonucleases at 37°C for 2 hours (see Table 1). Digestion was halted by incubation at 70°C for 15 minutes. The resulting nts were ligated into the expression vector, and E. coli or mammalian cells were transformed with the constructs by standard methods.
The DNA sequences of the Vh and Vl domains or of complete light chains of certain antibodies (GAL-FR21-mIgG1, GAL-FR22-mIgG2a and M048-D01-hIgG1-b) were synthesized by Geneart gene synthesis and gene optimizer technology for mammalian gene sion (life technologies, Grand Island, NY, USA). During the gene synthesis, the V domains on sequences coding for a mammalian signal peptide were fused with upstream Kozaki sequence. Flanking restriction cleavage sites were inserted on 5’ and 3’ ends of the synthesized DNA constructs. These restriction cleavage sites were used for the cloning of the Vh and Vl or the light chains into an expression vector which ned the coding constant regions of the IgG.
The expression vectors and the t constructs were digested according to cturer instructions with the respective restriction endonucleases at 37°C for 2 hours, to give compatible ends (see Table 1). Digestion was halted by incubation at 70°C for 15 minutes. The resulting fragments were ligated, and E. coli cells were transformed with the ligates. Plasmids obtained from these transformants were used to transform mammalian cells according to standard s.
Table1: Restriction cleavage sites for the cloning of Vh and Vl or of the light chain into IgG expression vectors Antibody Restriction ge site for the cloning of: VH VL or light chain* GAL-FR21-mIgG1 HindIII – BlpI XbaI – BsiWI GAL-FR22-mIgG2a HindIII – BlpI XbaI – BsiWI M048-D01-hIgG1# MfeI-BlpI BamHI – NotI* M048-D01-hIgG1-b I-BlpI XbaI – NotI* * Cloning of the complete light chain # Cloning of the PCR product into EcoRI-BlpI and NotI-digested expression vectors.
The antibodies were expressed transiently in mammalian cell culture, as described in Tom et al.
(Tom et al., Chapter 12 in Methods Express: Expression Systems, edited by Michael R. Dyson and Yves Durocher, Scion Publishing Ltd, 2007): for the expression of the anti-FGFR2 dies, for example M048-D01-hIgG1, GAL-FR21-mIgG1 and GAL-FR22-mIgG2a, HEK293 6E cells were transfected ently with a suitable CMV promoter-based expression plasmid. The cell culture scale was either up to 1.5 l in a shake flask or 10 l in a bag”. Expression took place at 37°C for 5-6 days in F17 medium (Invitrogen) supplemented with ne TN1 (Organotechnie) with 1% “FCS ultra low IgG” rogen) and 0.5 mM valproic acid.
Alternatively, the anti-FGFR2 antibodies, for example M048-D01-hIgG1-b, were expressed in a stably transfected Chinese hamster ovary (CHO) cell line. This was done using a one-vector system. Fermentation took place in bioreactors on a different scale in a fed-batch process.
The parental Fab fragment M048-D01 (Vh: SEQ ID NO: 21, Vl: SEQ ID NO: 22) belonging to the M048-D01-based antibodies from the phase display was sed as s: 20-50 ml of LB medium (admixed with 0.1 mg/ml ampicillin and 0.1% glucose) were inoculated with a preliminary culture of a corresponding E. coli clone, which contained the initial pBif vector, which was lacking the geneIII sequence, but which had had the M048-D01 Fab sequence cloned into it.
Production of the sFab was started by addition of 0.5 mM IPTG (final concentration). Incubation took place at 30°C overnight at 250 rpm.
B-3. Purification of the FGFR-2 antibodies The antibodies, for example 01-hIgG1, M048-D01-hIgG1-b, GAL-FR21-mIgG1 and GAL-FR22-mIgG2a, were obtained from the cell culture supernatants. The cell supernatants were clarified by centrifugation from cells. The cell supernatant was subsequently purified by affinity chromatography on a MabSelect Sure (GE Healthcare) chromatography . For this purpose, the column was equilibrated in DPBS pH 7.4 (Sigma/Aldrich), the cell supernatant was applied and the column was washed with around 10 column s of DPBS pH 7.4 + 500 mM NaCl.
The antibodies were eluted in 50 mM Na acetate pH 3.5 + 500 mM NaCl, and subsequently ed further by gel filtration chromatography on a Superdex 200 column (GE Heathlcare) in DPBS pH 7.4.
The al 01 Fab fragment expressed in E. coli was purified as follows: the E. coli cells were harvested by centrifugation and lysed by incubation at 4°C for 1 hour in lysis buffer (20% sucrose (w/v), 30 mM TRIS, 1 mM EDTA, pH 8.0, 1 mg/ml lysozyme (Sigma L-6876) and 2.5 U/ml Benzonase (Sigma E1014)). fter the same volume of PBS was added. After that, the ied supernatant was applied to Dynabeads for his-tag isolation (Invitrogen, 101-03D) and the mixture was swirled at 4°C for 2 hours. Thereafter the matrix was washed three times with buffer 1 (50 mM Na ate buffer, pH 7.4, 300 mM NaCl, 5 mM imidazole, 0.01% Tween 20).
Subsequently a single g step in buffer 2 was carried out (PBS admixed with 0.005% of Tween 20). Lastly, the Fabs were eluted with buffer E (10 mM Na phosphate buffer, pH 7.4, 300 mM NaCl, 300 mM imidazole) and concentrated using PBS buffer in Vivaspin 500 concentrators (cut-off 10 000, from GE, 2825).
B-4. Construction of the cross-reactivity profile of the M048-D01-based antibodies The cross-reactivity of the human antibodies M048-D01-hIgG1 and M048-D01-hIgG1-b of the present application was determined using the parental Fab fragment M048-D01 (comprising Vh: SEQ ID NO: 21 and Vl: SEQ ID NO: 22).
The M048-D01 Fab fragment was tested in an ELISA for binding to the various FGF receptor variants listed in Table 2.
WO 87716 Table 2: List of inant proteins used for establishing the cross-reactivity profile of the FGFR2 binders Protein Origin Cat. No. (RnD Systems) hFGFR2β-Fc (IIIb) Human 665-FR mFGFR2β-Fc (IIIb) Mouse 708-MF hFGFR2α-Fc (IIIb) Human 663-FR hFGFR2β-Fc (IIIc) Human 684-FR hFGFR1β-Fc (IIIc) Human 661-FR hFGFR1β-Fc (IIIb) Human 765-FR hFGFR3-Fc (IIIc) Human 766-FR hFGFR3-Fc (IIIb) Human 1264-FR hFGFR4-Fc Human 685-MF mFGFR2β-Fc (IIIc) Mouse 716-MF -Fc (IIIc) Mouse 710-MF hTRAIL-Fc Human 630-TR All of the variants took the form of Fc fusion proteins in r-free preparations. The proteins were biotinylated according to manufacturer instructions, using a 2-fold molar excess of biotin- LC-NHS (Pierce; Cat. No. 21347) and desalted using Zeba desalting columns (Pierce; Cat.
No. 89889).
For the ELISA, 96-well plates pretreated with streptavidin (Pierce, 15500) were loaded overnight at 4°C with 1 µg/ml biotinylated protein. Wells which had been loaded with biotinylated TRAIL- Fc served as a reference. The next day the plates were washed 3× with PBST (1 × PBS admixed with 0.05% Tween 20 (Sigma, P7949)), treated with ng buffer (PBST admixed with 3% BSA (Sigma A4503)), and again washed three times with PBST. 100 µl of the purified Fab (1 µg/ml) were added, and incubation was carried out at room temperature for 1 hour. After threefold washing with PBST, an HRP-coupled anti-hIgG (Fab-specific) (diluted 1:2500, Sigma, A0293) was added, and tion took place at room temperature for 1 hour. The colour reaction was activated by addition of 50 µl of TMB (Invitrogen, 2023) and halted after 5-15 s by addition of 50 µl of H2SO4 (Merck, 1120801000). The colour reaction was monitored at 450 nm in a plate reader (Tecan). The signal strengths of the wells containing TRAIL-Fc were used as ound values, and the signal-to-background ratios were calculated as summarized in Table 3.
Table 3: y of the ELISA data on the cross-reactivity of M048-D01 hFGFR2β -Fc -Fc -Fc -Fc -Fc -Fc -Fc (IIIb ) hFGFR2β (IIIc ) hFGFR2α (IIIb ) mFGFR2β (IIIb ) mFGFR2β (IIIc) β (IIIb ) β (IIIc) hFGFR3-Fc (IIIb) hFGFR3-Fc (IIIc) mFGFR3-Fc (IIIc) hFGFR4-Fc M048- +++ +++ +++ +++ +++ 0 0 0 0 0 0 Signal-to-background ratios: 0: <2; +: 2-3; ++: 3-5; +++: >5 As is evident from Table 2, the 01-based antibodies M048-D01-hIgG1 and M048-D01- hIgG1-b bind to human and murine FGFR2, and do so independently of whether the forms in question are alpha or beta isoforms, or are IIIb and IIIc splice forms. As is likewise apparent from the table, the M048-D01-based antibodies of the invention do not bind to FGFR1, FGFR3 and FGFR4.
B-5. Epitope mapping by means of CLIPS technology In order to test the binding characteristics of the M048-D01-based antibodies, an intensive epitope mapping operation was carried out on the basis of Pepscan’s proprietary “Chemically Linked peptides on Scaffolds” (CLIPS) Technologie (Timmerman et al., J. Mol. Recognit. 2007, - 99). In all, 8653 different CLIPS peptides were designed, from 15 amino acids to 30 amino acids in length, which cover linear, mational and discontinuous epitopes on the human FGFR2. The peptides were synthesized on peptide arrays. The human antibody M048-D01-hIgG1 was tested on the e arrays in an ELISA-based procedure. The peptides which gave the highest ELISA values were analysed to isolate common, similar amino acid ces.
In order to truct discontinuous epitopes of the target molecule, a y of structured peptides was synthesized. The CLIPS technology allows peptides to be structured in individual loops, double loops, triple loops, sheet-like loops, helix-like loops, and combinations of these structural elements: for this purpose, CLIPS templates are coupled to cysteine residues of the e arrays. For example, a 0.5 mM solution of the T2 CLIPS template 1,3- bis(bromomethyl)benzene was dissolved in ammonium bicarbonate (20 mM, pH 7.9)/acetonitrile (1:1 (v/v)). This solution was added to the peptide arrays. The CLIPS template bound to the side chains of 2 cysteines of the peptides present in the peptide arrays (455 well plate with 3 µl wells).
The peptide arrays were cautiously swirled in the solution for 30 to 60 minutes, with complete coverage by the solution. At the conclusion, the arrays were washed thoroughly with water in excess and were d in disrupt buffer (1% SDS, 0.1% beta-mercaptoethanol in PBS (pH 7.2)) at 70°C for 30 minutes in an ultrasound bath. The treatment in the ultrasound bath was uently repeated in water for a r 45 minutes. T3 CLIPS-bearing peptides were prepared in a r way.
The binding of the antibodies to each peptide was tested in a PEPSCAN-based ELISA (Sloostra et al., Molecular Diversity 1996, 1: 87-96). The peptide arrays were preincubated (1 h, 20°C) with % to 100% of binding buffer. The binding buffer consisted of 1% Tween 80, 4% horse serum and % ovalbumin (w/v) in solution in PBS. After a washing step, the peptide arrays were ted at 4°C overnight with primary antibody solution (1 to 5 µg/ml) in 1% Tween 80 in PBS. After a further washing step, the peptide arrays were incubated at 25°C for an hour in a 1/1000 dilution of an antibody peroxidase conjugate (anti-human-IgG) in 100% binder buffer. After a further g step, the peroxidase substrate 2,2’-azino-diethylbenzothiazolinesulphonate (ABTS) and 2 µl/ml 3% strength H2O2 were added. After an hour, the pment of colour was measured and was quantified using a CCD camera and an image processing .
The crude data obtained in this method are optical values which range from 0-3000 mAU (milli- absorption-units).
The result is that all the M048-D01-based antibodies bind on an epitope which consists of the 15 N-terminal residues of FGFR2 (1RPSFSLVEDTTLEPE 15 ). Analysis of 1257 CLIPS and linear peptides gave consistently high ELISA values for N-terminal peptides.
The N-terminal residues (1RPSFSLVEDTTLEPE 15 ) are present in all splice variants of human FGFR2, independently of alternative splicing in domain D3, which results in the IIIb and IIIc isoforms. The epitope is also present if the domain D1 is removed from the full-length FGFR2 (FGFR2 alpha, SEQ ID NO: 1) by splicing, resulting in the r beta-form of FGFR2 (SEQ ID NO: 2). In this case the epitope is situated ly before the domain D2.
A matter of particular interest is the fact that the N-terminal sequence is conserved in human, mouse, rat and rhesus monkey. This allows the broad inter-species cross-reactivity of the M048- D01 dies. The binding epitope of M048-D01-hIgG1 and M048-D01-hIgG1-b, two example dies of the present ion, is marked as the striped box in Figure 1.
B-6. General process for coupling to cysteine side chains The antibodies used in the coupling reactions were as follows: M048-D01-hIgG1 M048-D01-hIgG1-b Added to a solution of the corresponding antibody in PBS buffer in the concentration range between 1 mg/ml and 10 mg/ml were 3 equivalents of tris(2-carboxyethyl)phosphine hydrochloride (TCEP), in solution in PBS buffer, and the e was stirred at RT for 1 hour.
Subsequently, depending on the desired loading, between 2 and 10 equivalents of the maleimide precursor compound or halide precursor nd for coupling from the Intermediates 102, 103, 105-109, 111-114, 117-126, 128, 129, 132-146, 148-155, 157, 159-161, 166, 171, 175-177, 184, 189, 194-195, 199-201, 205, 209, 223-224, 226, 228-231, 236, 244 and 257 were added as a solution in DMSO. The amount of DMSO here ought not to exceed 10% of the overall volume.
The batch was stirred at RT for 60-120 s and then applied to PD 10 columns (Sephadex G- , GE Healthcare) equilibrated with PBS, and eluted with PBS buffer. Optionally a tration ure was carried out additionally by means of ultracentrifugation.
Normally, unless otherwise indicated, 5 mg of the corresponding antibody in PBS buffer were used for the ion and the subsequent coupling. ing purification via the PD10 column, this gave, in each case, solutions of the corresponding ADC in 3.5 ml of PBS buffer. The ular protein concentration indicated was then determined for these solutions. Furthermore, the loading of the antibody (drug/mAb ratio) was determined in ance with the methods described below.
This process was used to prepare the immunoconjugates represented in Examples 1, 3, 5-6, 8, 10- 12, 14, 15, 27 and 32.
In the structural formulae illustrated, the definition of AK1A and AK1B is as follows AK 1 1A = anti-FGFR2 antibody M048-D01-hIgG1 (partially reduced)- S§ AK 1B = anti-FGFR2 antibody M048-D01-hIgG1-b (partially reduced)- S§1 where §1 denotes the link with the succinimide group, and S stands for the sulphur atom of a cysteine residue of the partially d antibody.
B-7. General process for coupling to lysine side chains: The following antibodies were used in the ng reactions: M048-D01-hIgG1 01-hIgG1-b GAL-FR21-mIgG1 GAL-FR22-mIgG2a Added to a solution of the corresponding antibody in PBS buffer in the concentration range between 1 mg/ml and 10 mg/ml were, depending on the desired loading, between 2 and 5 equivalents of the precursor compound for coupling from the Intermediates 104, 110, 115, 116, 127, 130, 131, 147, 156, 158, 162, 169, 178, 185, 190, 202, 206, 210-216, 218, 219, 227, 233, 238, 240, 242, 245, 247a, 247b, 250, 251, 253, 255, 258 and 260-261 as a solution in DMSO. After minutes of stirring at RT, the same amount of precursor compound in DMSO was added again.
The amount of DMSO here ought not to exceed 10% of the overall volume. After a further minutes of stirring at RT, the batch was applied to PD 10 columns (Sephadex G-25, and eluted with PBS buffer. Optionally a concentration procedure was carried out additionally by means of ultrafiltration. If necessary, for more effective l of low molecular mass constituents, the concentration by ultrafiltration was repeated after ution with PBS buffer.
Normally, unless otherwise indicated, 5 mg of the corresponding antibody in PBS buffer were used for the coupling. Following purification via the PD10 column, this gave, in each case, solutions of the corresponding ADC in 3.5 ml of PBS buffer. The particular protein concentration ted was then ined for these solutions and the loading of the antibody (drug/mAb ratio) was determined in ance with the methods described below.
This process was used to prepare the immunoconjugates represented in Examples 2, 4, 7, 9, 13, 16- 17, 25, 26, 28-31 and 33-35.
In the structural formulae illustrated, the definition of AK2A, AK2B , AK2D and AK2E is as follows AK 2 2A = anti-FGFR2 antibody M048-D01-hIgG1 - NH§ AK 2 2B = anti-FGFR2 antibody M048-D01-hIgG1-b - NH§ AK 2D = anti-FGFR2 antibody GAL-FR21-mIgG1 - NH§2 AK 2E = anti-FGFR2 antibody GAL-FR22-mIgG2a - NH§2 where §2 denotes the link with the carbonyl group NH stands for the side chain amino group of a lysine residue of the antibody.
B-8. General process for ing ne s: µmol of the above-described maleimide precursor nds were taken up in 3 ml of DMF and admixed with 2.1 mg (20 µmol) of L-cysteine. The reaction mixture was d at RT for 2 hours, then concentrated under reduced pressure and subsequently purified by preparative HPLC.
In the structural formulae illustrated, the definition of Cys is as follows H N S §3 2 , where §3 denotes the link with the -toxophore unit.
B-9. General process for preparing lysine s: 10 µmol of the above-described active ester precursor compounds were taken up in 5 ml of DMF and d, in the presence of 30 µmol of N,N-diisopropylethylamine, with α-amino-protected L-lysine. The reaction mixture was stirred at RT for 2 hours, then concentrated under reduced pressure and subsequently purified by ative HPLC. The tive group was then removed by known methods.
Further purification and characterization of the conjugates of the invention After on had taken place, in certain cases the reaction mixture was concentrated, by ultrafiltration, for example, and then desalted and purified by means of chromatography, for example using a ex® G-25. Elution took place with, for example, phosphate-buffered saline (PBS). The solution was subsequently subjected to sterile filtration and freezing. An alternative option is to lyophilize the conjugate.
B-10. ination of the toxophore loading The toxophore loading of the resultant ons of the conjugates described in the working examples, in PBS buffer, was determined as follows: The toxophore loading of lysine-linked ADCs was determined by mass-spectrometric determination of the molecular weights of the individual ate species. In this case, to start with, the antibody conjugates were deglycosylated by means of PNGaseF, and the sample was acidified and, following HPLC separation, was analysed by mass spectrometry using an ESI-MicroTofQ (Bruker Daltonik). All of the spectra were added via the signal in the TIC (Total Ion Chromatogram), and the molecular weight of the various conjugate s was calculated on the basis of MaxEnt Deconvolution. Following signal integration of the different species, the DAR (Drug/Antibody Ratio) was then calculated.
For protein identification, in addition to the molecular weight determination, a tryptic digestion was carried out after osylation and/or denaturing, and this digestion, after denaturing, reduction and derivatization, med the ty of the protein on the basis of the tryptic peptides ed.
The toxophore loading of cysteine-linked conjugates was determined via reversed-phase chromatography of the reduced and denatured ADC. The ADC solution (1 mg/mL, 50 µL) was admixed with guanidinium hydrochloride (GuHCl) (28.6 mg) and with a solution of DL-dithiothreitol (DTT) (500 mM, 3 µL). The mixture was ted at 55°C for an hour and analysed by HPLC.
The HPLC analysis was carried out on an Agilent 1260 HPLC System with detection at 220 nm.
The column used was a r Laboratories PLRP-S Polymeric Reversed Phase column (catalogue number PL1912-3802) (2.1 × 150 mm, 8 µm particle size, 1000 Å) with a flow rate of 1 mL/min, using the following gradient: 0 min, 25% B; 3 min, 25 % B; 28 min, 50% B. Eluent A consisted of 0.05% trifluoroacetic acid (TFA) in water, eluent B of 0.05% trifluoroacetic acid in acetonitrile.
The peaks detected were assigned by retention time comparison with the light chain (L0) and the heavy chain (H0) of the unconjugated antibody. Peaks which were detected ively in the conjugated sample were assigned to the light chain, with a toxophore (L1), and to the heavy chains, with one, two and three toxophores (H1, H2, H3).
The average loading of the dy with toxophores was determined from the peak areas determined by integration as follows: the light-chain loading was calculated as the sum of the ore number weighted integration results of the peaks belonging to the light chains d by the sum of the singularly weighted ation results of the peaks belonging to the light .
The heavy-chain loading was calculated as the sum of the toxophore number weighted integration s of the peaks belonging to the heavy chains divided by the sum of the singularly weighted integration results of the peaks belonging to the heavy chains. The average drug load s therefrom as the twofold sum of light-chain loading and heavy-chain loading. In certain individual cases it may be impossible exactly to determine the toxophore loading, owing to co-elutions of certain peaks.
B-11. Testing of the antigen binding of the ADC The binding capacity of the binder to the target molecule was tested after coupling had taken place.
The skilled worker knows of diverse methods for achieving this – for example, the affinity of the conjugate can be tested by means of ELISA technology or surface plasmon resonance analysis (BIAcore™ measurements). The conjugate concentration can be measured by the skilled person using common methods – for example, for antibody conjugates, by means of protein determination 2012/075277 (see also Doronina et al.; Nature Biotechnol. 2003; 21:778-784 and Polson et al., Blood 2007; 1102:616-623).
Working es - immunoconjugates Example 1 AK O 1A H C CH H C 3 3 3 CH N O 3 H H N N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 Coupling here was carried out using 30 mg of M048-D01-hIgG1 in PBS and following the Sephadex purification the batch was concentrated by ultracentrifugation, re-diluted with PBS and concentrated again.
Protein concentration: 15.5 mg/ml Drug/mAb Ratio: 3.7 Example 2 H C CH3 H C 3 3 CH 3 N O H H N AK2A N N O N N O O O CH3 O CH O O 3 CH3 H C CH3 CH 3 3 Coupling here was carried out using 32 mg of 01-hIgG1 in PBS and following the Sephadex purification the batch was concentrated by ultracentifugation, re-diluted with PBS and concentrated again.
Protein tration: 11.7 mg/ml Drug/mAb Ratio: 3.9 WO 87716 Example 3 H C CH H C 3 3 3 CH 3 N O O H H N AK O O N N O 1A N O N N O O CH O CH O O H C 3 3 3 O H C CH CH 3 3 3 ng here was carried out using 30 mg of M048-D01-hIgG1 in PBS and following the Sephadex purification the batch was concentrated by ultracentrifugation, re-diluted with PBS and concentrated again.
Protein concentration: 12.5 mg/ml Drug/mAb Ratio: 3.7 Example 4 H C CH H3C 3 3 CH3 N O H AK2A H N O O N N O O N N O O O CH O CH O O 3 3 CH3 H C CH CH3 3 3 H n Coupling here was carried out using 2 mg of M048-D01-hIgG1 in PBS and following the ex purification the batch was concentrated by ultracentifugation and re-diluted with PBS.
Protein concentration: 1.92 mg/ml Drug/mAb Ratio: 3.7 Example 5 3 O AK1A O H C CH H C 3 3 3 CH N O O 3 H H H N N N N N O N N N H O O O O CH O CH O O 3 3 H C H C CH CH3 3 3 3 Coupling here was carried out using 3 mg of M048-D01-hIgG1 in PBS and following the Sephadex purification the batch was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 2.54 mg/ml Drug/mAb Ratio: 3.1 Example 6 1A O H C CH H C 3 3 3 CH 3 NH O O H 2 H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 Coupling here was carried out using 4 mg of M048-D01-hIgG1 in PBS and following the Sephadex purification the batch was trated by entrifugation and uted.
Protein concentration: 1.53 mg/ml Drug/mAb Ratio: 2.7 Example 7 H C CH H C 3 3 3 CH3 NH O 2 AK2B H N O O N N O O N N O O O CH3 O CH3 O O CH H C CH CH 3 3 3 3 Coupling here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and following the Sephadex purification the batch was trated by ultracentrifugation and re-diluted.
Protein concentration: 1.28 mg/ml Drug/mAb Ratio: 6.1 Example 8 H C CH H C 3 3 3 CH O 3 N O H H N AK O O N N O 1B N O N N O O CH O CH O O H C 3 3 3 O H C CH CH 3 3 3 Coupling here was d out using 5 mg of M048-D01-hIgG1-b in PBS and following the Sephadex purification the batch was concentrated by ultracentifugation and uted.
Protein concentration: 1.26 mg/ml Drug/mAb Ratio: 3.5 Example 9 H C CH3 H3C 3 CH N O 3 H N AK2B N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 H C 3 3 3 Coupling here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and following the Sephadex purification the batch was concentrated by ultracentifugation and re-diluted.
Protein concentration: 1.28 mg/ml Drug/mAb Ratio: 6.1 Example 10 H C CH H C 3 3 3 CH N O 3 O H H H H N N N N N O AK N N N 1B O O O CH O CH O O 3 3 CH O H C CH CH 3 3 3 3 Coupling here was d out using 5 mg of M048-D01-hIgG1-b in PBS and following the Sephadex purification the batch was concentrated by ultracentrifugation and re-diluted. n concentration: 1.36 mg/ml Drug/mAb Ratio: 4.4 Example 11 AK1B O O H C CH H C 3 3 3 CH 3 N O H H N N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 H C 3 3 3 3 Coupling here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and following the Sephadex purification the batch was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 1.27 mg/ml Drug/mAb Ratio: 4.8 Example 12 O CH CH AK1B 3 3 O H C CH H C 3 3 3 CH N O HN O O 3 H H N N N O N N O N N N H O O O O CH O CH O CH O O CH 3 3 3 H C CH CH 3 3 3 3 Coupling here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and following the Sephadex purification the batch was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 1.48 mg/ml Drug/mAb Ratio: 4.0 Example 13 H C CH H C O 3 3 O 3 CH 3 H N H N AK2B N N N N N O O O CH O CH O O CH 3 H C CH 3 3 3 CH ng here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and following the Sephadex purification the batch was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 1.21 mg/ml Drug/mAb Ratio: 1.5 e 14 AK1B 3 O H C CH H C O 3 3 3 CH O 3 H N H N N N N N N N O O O CH O CH O O CH H C CH 3 3 3 3 CH ng here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and the batch, following Sephadex purification, was trated by ultracentrifugation and re-diluted.
Protein concentration: 1.27 mg/ml Drug/mAb Ratio: 2.7 Example 15 AK CH CH 3 3 1B O O H C CH H C 3 3 3 CH N O HN O O 3 H H N N N O N N O N N N H O O O O CH O CH3 O CH O O 3 3 CH3 H C CH CH 3 3 3 Coupling here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and the batch, following Sephadex purification, was concentrated by entrifugation and re-diluted with PBS.
Protein concentration: 1.37 mg/ml Drug/mAb Ratio: 3.9 Example 16 H3C CH H C 3 3 CH O 3 N H N AK2D N N O N N O O O CH O CH3 O O 3 CH H3C CH CH 3 3 3 H n Coupling here was carried out using 4 mg of GAL-FR21-mIgG1 in PBS and the batch, following Sephadex purification, was trated by ultracentrifugation and re-diluted with PBS.
Protein concentration: 1.63 mg/ml Drug/mAb Ratio: 8.7 Example 17 H C CH3 H3C 3 CH N O 3 H N AK2E N N O N N O O O CH O CH3 O O 3 CH H C CH CH 3 3 3 3 Coupling here was carried out using 4 mg of GAL-FR22-mIgG2a in PBS and the batch, following Sephadex cation, was trated by ultracentrifugation and re-diluted with PBS.
Protein concentration: 1.60 mg/ml Drug/mAb Ratio: 8.1 Example 18 N-(6-{[(5S)Aminocarboxypentyl]amino}oxohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropanyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide trifluoroacetate H C CH H C 3 3 3 CH 3 N O H H H N H N N N N 2 O N N O O COOH O CH3 O CH O O 3 CH H C CH CH 3 3 3 3 x H N H .5 mg (15 µmol) of Intermediate 210 were taken up in 5 ml of DMF and d with 4.4 mg (18 µmol) of N2-(tert-butoxycarbonyl)-L-lysine and also 7.7 µL (44 µmol) of N,N- diisopropylethylamine. The reaction mixture was stirred at RT overnight and then concentrated under reduced pressure. The residue was subsequently purified by preparative HPLC. This gave 14 mg (81% of theory) of the protected intermediate of the title compound, which was subsequently taken up in 1 ml of dichloromethane and deprotected with 1 ml of trifluoroacetic acid. The batch was concentrated and, following lyophilization of the residue from acetonitrile/water (1:1), 15 mg (97% of ) of the title compound were obtained.
HPLC (Method 12): Rt = 1.8 min; LC-MS (Method 1): Rt = 0.79 min; MS (ESIpos): m/z = 1083 (M+H)+.
Example 19 N-(6-{[(5S)Aminocarboxypentyl]amino}oxohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S) 2-[(1R,2R){[(1S)carboxy(1H-indolyl)ethyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide H C CH3 H C 3 3 CH3 OH O H H H N H N N N N 2 O N N O O COOH O CH3 O CH O O 3 CH H3C CH3 CH 3 40 mg (40 µmol) of ediate 227 were taken up in 5 ml of DMF and admixed with 11.5 mg (40 µmol) of N2-[(benzyloxy)carbonyl]-L-lysine and also 13 µL (80 µmol) of N,N- diisopropylethylamine. The reaction mixture was stirred at RT overnight, then concentrated under reduced pressure and subsequently purified by preparative HPLC. This gave 32.5 mg (70% of theory) of the ted intermediate of the title compound.
This 32.5 mg of the ediate were dissolved in 10 ml of methanol and, following addition of 2 mg of 10% palladium on activated , were hydrogenated under standard hydrogen pressure at RT for 30 s. The catalyst was then removed by filtration and the solvent was removed under reduced pressure. lization of the residue from dioxane/water 1:1 gave 26 mg (99% of theory) of the title compound.
HPLC (Method 12): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.76 min; MS (ESIpos): m/z = 1014 (M+H)+.
Example 20 N-[(18S)Aminocarboxyoxo-3,6,9-trioxaazaoctadecyl]-N-methyl-L-valyl-N- S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl)oxopropan yl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptan yl]-N-methyl-L-valinamide trifluoroacetate H C CH H C 3 3 3 CH3 N O H H H N H2N N O O N N O O N N O O COOH O CH3 O CH O O 3 CH H3C CH CH3 3 x CF3COOH N 3.5 mg (3 µmol) of Intermediate 202 were taken up in 2 ml of DMF and admixed with 0.8 mg (3 µmol) of N2-(tert-butoxycarbonyl)-L-lysine and also 1.6 µL (10 µmol) of N,N- diisopropylethylamine. The reaction mixture was stirred at RT overnight and then concentrated under reduced pressure. The residue was taken up in acetonitrile/water (1:1), brought to a pH of 2 with trifluoroacetic acid and then purified by preparative HPLC. This gave 1 mg (25% of theory) 2012/075277 of the protected intermediate of the title compound, which was subsequently taken up in 500 µl of dichloromethane and deprotected with 500 µl of trifluoroacetic acid. The batch was concentrated and, following lypophilization of the residue from acetonitrile/water (1:1), 1 mg (89% of theory) of the title compound was obtained.
HPLC (Method 12): Rt = 1.9 min; LC-MS (Method 1): Rt = 0.82 min; MS (ESIpos): m/z = 1173 (M+H)+.
Example 21 N-(4-{2-[6-(3-{[(2R)Aminocarboxyethyl]sulphanyl}-2,5-dioxopyrrolidinyl)hexanoyl] hydrazino}oxobutyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)amino dolyl)oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl} methoxymethyloxoheptanyl]-N-methyl-L-valinamide O H C CH H3C 3 3 CH 3 NH2 O O H H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 mg (10 µmol) of Intermediate 157 were taken up in 5.2 ml of DMF and admixed with 2.28 mg (20 µmol) of L-cysteine. The on e was stirred at RT for 2 hours, then concentrated under reduced pressure and subsequently purified by preparative HPLC. This gave 5.8 mg (48% of theory) of the title compound.
HPLC (Method 5): Rt = 1.45 min; LC-MS (Method 1): Rt = 0.74 min; MS (ESIpos): m/z = 1184 (M+H)+.
Example 22 N-(4-{2-[6-(3-{[(2R)Aminocarboxyethyl]sulphanyl}-2,5-dioxopyrrolidin yl)hexanoyl]hydrazino}oxobutyl)-N-methyl-L-valyl-N-[(3R,4S,5S){(2S)[(1R,2R) {[(1S)carboxy(1H-indolyl)ethyl]amino}methoxymethyloxopropyl]pyrrolidin yl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide Cys 3 O H C CH3 H C 3 3 CH OH O O 3 H H H N N N N N O N N N H O O O O CH O CH O O 3 3 CH H C CH3 CH 3 3 3 mg (10 µmol) of Intermediate 113 were taken up in 5.2 ml of DMF and admixed with 2.28 mg (20 µmol) of eine. The reaction mixture was stirred at RT for 2 hours, then concentrated under reduced pressure and subsequently purified by preparative HPLC. This gave 6 mg (54% of theory) of the title compound.
HPLC (Method 5): Rt = 1.5 min; LC-MS (Method 1): Rt = 0.77 min; MS s): m/z = 1185 .
Example 23 N-[6-(3-{[(2R)Aminocarboxyethyl]sulphanyl}-2,5-dioxopyrrolidinyl)hexyl]-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(1S)carboxy(1H-indolyl)ethyl]amino} methoxymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl- L-valinamide Cys 3 O H C CH H C 3 3 3 CH 3 OH O H H N N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 mg (10 µmol) of Intermediate 124 were taken up in 4 ml of DMF and admixed with 2.5 mg (20 µmol) of L-cysteine. The reaction mixture was stirred at RT for 2 hours, then concentrated under reduced pressure and subsequently purified by preparative HPLC. This gave 7.2 mg (64% of theory of the title compound.
HPLC (Method 5): Rt = 1.6 min; LC-MS (Method 1): Rt = 0.8 min; MS (ESIpos): m/z = 1071 (M+H)+.
Example 24 N-[6-(3-{[(2R)Aminocarboxyethyl]sulphanyl}-2,5-dioxopyrrolidinyl)hexyl]-N-methyl-L- valyl-N-[(3R,4S,5S){(2S)[(1R,2R){[(2S)(1H-indolyl)(1,2-oxazinanyl) oxopropanyl]amino}methoxymethyloxopropyl]pyrrolidinyl}methoxymethyl oxoheptanyl]-N-methyl-L-valinamide Cys 3 O H C CH H C 3 3 3 CH N O 3 H N N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 mg (10 µmol) of Intermediate 125 were taken up in 4 ml of DMF and admixed with 2.4 mg (20 µmol) of L-cysteine. The reaction mixture was stirred at RT for 2 hours, then concentrated under reduced re and subsequently purified by preparative HPLC. This gave 7.7 mg (69% of theory of the title compound.
HPLC (Method 5): Rt = 1.7 min; LC-MS (Method 2): Rt = 1.91 min; MS (ESIpos): m/z = 1140 (M+H)+. e 25 H C CH H C CH O 3 3 3 N O 3 H H N S N N N O 2B S N N O O O CH O CH O O H C 3 3 H C CH CH 3 3 3 3 H n Coupling here was d out using 5 mg of M048-D01-hIgG1-b in PBS and the batch, following Sephadex purification, was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 1.27 mg/ml Drug/mAb Ratio: 1.0 Example 26 H C CH H C 3 3 3 CH 3 N O H H N AK2B N N O N N O O O CH O CH O O 3 3 CH3 H C CH CH3 3 3 Coupling here was carried out using 35 mg of M048-D01-hIgG1-b in PBS and the batch, following Sephadex cation, was concentrated by ultracentrifugation, re-diluted with PBS and concentrated again.
Protein concentration: 11.60 mg/ml Drug/mAb Ratio: 3.7 e 27 AK1B O H C CH H C 3 3 3 CH 3 N O H H N N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 Coupling here was carried out using 35 mg of M048-D01-hIgG1-b in PBS and the batch, following Sephadex purification, was concentrated by ultracentrifugation, uted with PBS and concentrated again.
Protein concentration: 11.7 mg/ml Drug/mAb Ratio: 4.2 Example 28 H C CH H C 3 3 3 CH N O 3 H AK2B H N N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 Coupling here was d out using 5 mg of M048-D01-hIgG1-b in PBS and the reaction e, following Sephadex purification, was concentrated by ultracentrifugation and re-diluted with PBS.
Protein concentration: 1.31 mg/ml Drug/mAb Ratio: 3.7 Example 29 H C CH H C 3 CH 3 3 N O 3 H H N 2B O O N N O O N N O O O CH O CH O O H C 3 3 H CH CH 3 3 3 3 Coupling here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and the reaction mixture, following Sephadex purification, was concentrated by ultracentrifugation and re-diluted with PBS.
Protein concentration: 1.53 mg/ml Drug/mAb Ratio: 1.3 Example 30 H C CH H C 3 3 3 CH N O O O 3 H H N N N N O AK N N N 2B O O 3 O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 H n Coupling here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and the batch, following Sephadex purification, was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 1.61 mg/ml Drug/mAb Ratio: 3.9 Example 31 H C CH H3C O 3 3 CH O 3 H N AK2B H N O O N N N O N N O O O CH3 O CH O O CH H C CH3 3 3 CH Coupling here was d out using 5 mg of 01-hIgG1-b in PBS and the reaction mixture, following Sephadex purification, was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 1.32 mg/ml Drug/mAb Ratio: 1.7 Example 32 H3C CH H C CH O 3 O 3 3 O H N H N O N N AK1B O N N O N N O O CH O CH3 O O CH3 O H C CH 3 3 CH3 Coupling here was d out using 5 mg of M048-D01-hIgG1-b in PBS and the reaction mixture, following Sephadex purification, was concentrated by ultracentrifugation and re-diluted. n concentration: 1.12 mg/ml Drug/mAb Ratio: 0.2 Example 33 H C CH H C 3 3 3 CH 3 N O H AK2B H N N N O N N O O O CH O CH3 O O 3 CH3 H3C CH CH 3 3 ng here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and the reaction mixture, following Sephadex purification, was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 1.29 mg/ml Drug/mAb Ratio: 5.5 Example 34 H C CH H C 3 3 3 CH O 3 H N AK N N N N O O O CH O CH O O CH 3 H C CH H3C 3 3 3 Coupling here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and the reaction mixture, following Sephadex purification, was concentrated by entrifugation and re-diluted.
Protein tration: 1.04 mg/ml Drug/mAb Ratio: 0.5 Example 35 H C CH H C 3 3 3 CH 3 N O H H N AK2B O O N N O O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 ng here was carried out using 5 mg of M048-D01-hIgG1-b in PBS and the on mixture, following Sephadex purification, was concentrated by ultracentrifugation and re-diluted.
Protein concentration: 1.66 mg/ml Drug/mAb-Ratio: 3.4 Example 36 N-(6-{[(5S)Aminocarboxypentyl]amino}oxohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R){[(2S)(4-hydroxyphenyl)(1,2-oxazinanyl)oxopropanyl]amino} ymethyloxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L- valinamide trifluoroacetate H C CH H C 3 3 3 CH3 N O H H H N H N N N N 2 O N N O O COOH O CH O CH3 O O 3 CH H C CH CH 3 3 3 3 x H OH 8 mg (8 µmol) of Intermediate 242 were taken up in 3 ml of DMF and admixed with 2.9 mg (12 µmol) of N2-(tert-butoxycarbonyl)-L-lysine and also 2.7 µL (16 µmol) of N,N- diisopropylethylamine. The reaction mixture was stirred at RT overnight, then admixed again with the same amounts of N2-(tert-butoxycarbonyl)-L-lysine and N,N-diisopropylethylamine, and stirred at RT for a further 4 hours. The batch was subsequently concentrated under reduced pressure. The residue was then purified by preparative HPLC. Lyophilization from acetonitrile/water gave 6.5 mg (72% of theory) of the protected intermediate of the title compound, which was subsequently taken up in 5 ml of dichloromethane and deprotected with 0.75 ml of trifluoroacetic acid. The batch was concentrated, and lyophilization of the e from dioxane/water gave 5 mg (76% of theory) of the title compound.
HPLC (Method 12): Rt = 1.7 min; LC-MS (Method 1): Rt = 0.69 min; MS (ESIpos): m/z = 1059 (M+H)+.
Example 37 N-(6-{[(5S)Aminocarboxypentyl]amino}oxohexyl)-N-methyl-L-valyl-N-[(3R,4S,5S) {(2S)[(1R,2R){[(1S)carboxy(4-hydroxyphenyl)ethyl]amino}methoxymethyl oxopropyl]pyrrolidinyl}methoxymethyloxoheptanyl]-N-methyl-L-valinamide trifluoroacetate H C CH3 H C 3 3 CH3 OH O H H H N H N N N N 2 O N N O O COOH O CH O CH O O 3 3 CH H3C CH3 CH 3 x CF3COOH OH 38 mg (41 µmol) of Intermediate 248 were first converted into the N-hydroxysuccinimide ester. 72 mg of the crude product obtained were taken up in 5 ml of DMF and admixed with 24 mg (100 µmol) of N2-(tert-butoxycarbonyl)-L-lysine and 23 µL of isopropylethylamine. The reaction mixture was stirred at RT overnight, and then d again with 16 mg of N2-(tert- butoxycarbonyl)-L-lysine and 12 µL of N,N-diisopropylethylamine, and subsequently treated in an ound bath for a further 2 hours. The batch was then concentrated under reduced pressure and the residue was purified by preparative HPLC. Lyophilization from acetonitrile/water gave 20 mg (50% of theory) of the protected intermediate of the title compound. mg (12 µmol) of this intermediate were subsequently taken up in 3 ml of dichloromethane and admixed with 1 ml of trifluoroacetic acid. After 40 minutes of stirring at RT, a r 1.5 ml of oroacetic acid were added and the batch was treated in an ultrasound bath for 1 hour.
Thereafter the batch was concentrated, and lyophilization of the residue from dioxane/water gave 13 mg (90% of ) of the title compound.
HPLC (Method 12): Rt = 1.5 min; LC-MS (Method 1): Rt = 0.68 min; MS (ESIpos): m/z = 990 (M+H)+.
C: Evaluation of biological activity: The biological effect of the compounds of the invention was demonstrated by the assays described below: C-1. Identification of tumour cells with different FGFR2 levels on the cell surface To determine the amounts of FGFR2 available for the antibody on the cell surface, the cell binding of the FGFR2 antibody on different tumour cell lines was analysed in flow cytometry. The cell lines below were used for the experiments. The information on mutations and copy number of the FGFR2 comes from the Sanger Center Genome Project: - SNU-16 human stomach carcinoma cells, FGFR2 gene amplification (copy number 14; ATCC- CRL-5974, RPMI 1640 rom FG1215) + 10% FCS - KatoIII human stomach carcinoma cells, FGFR2 gene amplification (copy number 14 ATCCTCP-1008 ; Iscove’s (Biochrom FG0465) + 20% FCS - SUM52-PE human breast cancer cells, FGFR2 gene amplification (copy number 14; Asterand Lot No.: 1-6004; Ham’s F12 (Biochrom FG0815) + 5% FCS + 10 mM Hepes buffer + 1 µg/ml hydrocortisone + 5 µg/ml insulin - MFM-223 human breast cancer cells, FGFR2 gene amplification (copy number 14; ECACC- 98050130, MEM Earle rom F0315) + 10% FCS + 2mM glutamine - NCI-H716 human colorectal oma cells, FGFR2 gene amplification (copy number: 8; ATCC-CCL-251; RPMI 1640 (Biochrom FG1215) + 10% FCS) - MDA-MB-231 human breast cancer cells (no FGFR2 gene amplification, copy number 3; ATCC-HTB-26, DMEM/HAM´s F12 rom FG4815) + 10% FCS For the experiments, adherent cells were washed twice with PBS (without calcium (Ca2+ ) and magnesium (Mg2+ ) ions and detached using enzyme-free, PBS-based ce ll dissociation buffer (Invitrogen). Approximately 1 × 105 cells per well were ded in FACS buffer (PBS without Ca 2+ and Mg2+ with 3% FCS (Biochrom)), followed by fugation (250 g, 5 min, 4°C), and the supernatant was ded. The cells were resuspended in antibody dilutions (5 µg/ml in 80 µl) in FACS buffer, and incubated on ice for 1 hour. Next, the cells were washed once with 100 µl of cold FACS buffer and 80 µl of 1:150 diluted secondary antibodies (PE-coupled goat anti-mouse IgG, Jackson Immuno ch #115164 for GAL-FR21-mIgG1 and for GAL-FR22-mIgG2a, and pled goat anti-human IgG, Dianova #109098 for M048-D01-hIgG1) were added.
Following incubation on ice for 1 hour, the cells were again washed with cold FACS buffer, resuspended in 100 µl of FACS , and analysed in an FACS array flow cytometer (BD Biosciences). The s were calculated as the geometric average value of the cell population detected with FGFR2 dy, minus the background fluorescence, which was measured by incubating the cell population only with the ary antibody. The values were analysed by the following system: geometric average value FGFR2 antibody minus geometric average value only secondary antibody > 10 +, > 100 ++, > 1000 +++, > 10000 ++++, -: no signal. Values in the vicinity of the category limits are marked with ().
The binding of FGFR2 dies to tumour cells is indicated in Table 4: Table 4 M048-D01-hIgG1 GAL-FR21-mIgG1 GAL-FR22-mIgG2a 3 +++ +++ +++ SNU-16 ++(+) ++(+) ++ MDA-MB-231 - nd 1) nd1) 1) nd: not ined The FGFR2 antibodies detect FGFR2 on the cell surface of MFM-223 and SNU-16 cancer cells.
C-2. Determination of the cytotoxic effect of the ADCs directed against FGFR2 The cytotoxic effect of the FGFR2 ADCs was determined on various cell lines with different expression quantities of FGFR2, as follows: The cells were cultured according to a standard method, using the growth media indicated under C-1. For the implementation, the cells were detached with a solution of trypsin (0.05%) and EDTA (0.02%) in PBS (Biochrom AG #L2143), pelletized, resuspended in culture medium, counted, and seeded into a 96-well culture plate with a white base (Costar #3610) (at 75 µl/well, following cell counts per well: SNU-16: 3000; MFM-223: 7000; MDA-MB-231: 4000; SUM52-PE: 3000; NCIH716 : 3000; KatoIII: 3000) and incubated in an incubator at 37°C under 5% carbon dioxide. After 24 hours, the antibody-drug ates in 25 µl of culture medium (four-fold concentration) were applied to the cells, giving final antibody-drug conjugate concentrations of 3 × 10-7 M to 3 × 10-11 M on the cells (triplicates). The cells were then incubated in an incubator at 37°C and 5% carbon e. In a parallel plate, the cell vitality at the beginning of drug ent (day 0) was determined using the Cell Titer Glow Luminescent Cell Viability Assay (Promega #G7573 and #G7571). For this, 100 µl of the substrate were added per cell batch, and the plates were then d with aluminium foil, shaken at 180 rpm with a plate shaker for 2 minutes, left to stand on the laboratory bench for 8 minutes, and then measured using a luminometer r X2, Perkin Elmer). The substrate detects the ATP content in the living cells, producing a luminescence signal whose level is directly proportional to the vitality of the cells. After 72 hours of incubation with the antibody-drug conjugates, the ty was determined in these cells as well, using the Cell Titer Glow Luminescent Cell Viability Assay as described above. From the data ed, the IC50 of growth inhibition was calculated in ison to that on day 0, using the laboratory software MTS (developed by Schering AG and Bayer Business Services 1999-2009) on the basis of a 4-parameter adaptation.
Table 5 below lists the IC50 values1) of representative working examples from this assay: Table 5 MDA- SUM52-PE, NCI-H716, KatoIII, SNU- MFM- MB-231, IC50, 72 hr IC50, 72 hr IC50, 72 hr 16, 223, IC50, 72 [nM] [nM] [nM] IC50, 72 IC50, 72 hr Example hr [nM] hr [nM] [nM] 1 0.214 0.0907 > 300 2 1.19 0.86 > 300 3 0.15 0.195 > 300 4 0.567 1.47 > 300 0.496 4.00 > 300 6 0.497 6.85 > 300 7 0.209 0.628 > 300 8 2.06 8.67 256 9 2.05 4.78 230 0.519 10.3 > 300 11 0.614 0.581 80.9 12 0.389 0.145 > 300 13 0.254 0.431 6.61 14 0.914 1.91 67.5 16.6 16.9 > 300 16 < 0.03 0.0788 > 300 17 0.0616 0.278 > 300 26 0.455 na > 248 < 0.097 4.69 na 27 0.159 0.109 > 248 < 0.053 0.517 0.198 28 0.277 na na 0.101 na na 29 1.70 na na 0.106 na na 1.84 9.16 > 248 0.244 na na 31 5.88 na na na na na 32 > 300 na na na na na 33 8.50 na 128 0.104 na 0.920 34 7.41 na > 248 3.37 na na na na > 248 0.105 na 0.523 1) The activity data reported relate to the working examples described in the present experimental section, with the drug/mAB ratios indicated. The values may ly deviate for different drug/mAB ratios.
Working Example 1 inhibited the proliferation of the SNU-16 and 3 cancer cell lines which expresses FGFR2 at the cell surface, with an IC50 in the omolar concentration range.
Working Example 1 inhibited the proliferation of the MDA-MB-231 cancer cell line which do not s FGFR2 at the cell surface, with an IC50 of 300 nM. As is apparent from the data, all tested dy-drug conjugates (Working Examples 1-31, 33-35) selectively inhibit the proliferation of FGFR2-expressing cancer cell lines 6, MFM-223, SUM52-PE, KatoIII or NCI-H716).
C-3. Determination of the effect on tubulin polymerization Cancer cells are denatured cells which frequently lead to the formation of tumours also as a result of sed cell division. Microtubuli form the spindle fibres of the spindle apparatus and are an essential constituent of the cell cycle. The ted construction and breakdown of microtubuli allows the precise division of the chromosomes among the daughter cells, and constitutes a uously dynamic process. Disruption to this dynamic process results in incorrect cell on and ultimately in cell death. The increased cell division of cancer cells, however, also makes them particularly sensitive towards spindle fibre poisons, which tute a fixed constituent of chemotherapy. Spindle fibre poisons such as paclitaxel or epothilone lead to a sharply increased polymerization rate of the microtubuli, while vinca alkaloids or else monomethylauristatin E (MMAE) lead to a sharply reduced polymerization rate of the microtubuli. In both cases, the necessary dynamism of the cell cycle is critically disrupted.
Tubulin polymerization was igated using the "Fluorescence-based Microtubule Polymerisation Assay Kit" from Cytoskeleton (Denver, Colorado, USA; order number: BK011).
With this assay, GTP is added to unpolymerized tubulin, allowing polymerization to take place spontaneously. The assay is based on the binding of the fluorophore 4',6-diamidinophenylindole (DAPI) to tubulin. Free and bound DAPI can be differentiated on the basis of different emission spectra. Since DAPI exhibits a significantly high affinity for polymerized tubulin in ison to lymerized tubulin, the tubulin polymerization can be followed via the increase in the fluorescence of bound DAPI fluorophores.
For the implementation of this assay, the compounds of the invention, in solution in DMSO, were diluted from their initial concentration of 10 mM to 1 µM in water. In on to the buffer control, paclitaxel, with a polymerization-increasing effect, and vinblastin, with a polymerization inhibiting effect, were run additionally as assay controls. Measurement was carried out using 96- well plates with a half base area. The kinetics of the tubulin polymerization were monitored in a Fluorimeter at 37°C for 1 hour. The excitation wavelength was 355 nm, and emission was red at 460 nm. For the region of linear increase within the first 10 minutes, a calculation was made of the change in fluorescence per minute (∆F/min), which represents the polymerization rate of the microtubuli. The potency of the test nces was quantified on the basis of their respective ion of the polymerization rate. Table 6 below gives data for the influence of representative working examples on tubulin polymerization.
Table 6: de of tubulin polymerization by selected examples of toxophore variants.
Working example Concentration of Tubulin polymerization in the presence of toxophore in [%]. toxophore Tubulin polymerization rate at [µM] 1 µM MMAF set at 100% MMAF 1 100 MMAF 10 34 MMAF 100 0 18 1 45 18 10 1 19 1 80 19 10 14 1 60 10 0 Working example Concentration of Tubulin polymerization in the presence of toxophore in [%]. toxophore Tubulin polymerization rate at [µM] 1 µM MMAF set at 100% 21 1 88 21 10 25 22 1 109 22 10 27 24 1 121 24 10 35 36 1 88 36 10 21 37 1 90 37 10 17 The MMAF toxophore and the working examples inhibit n polymerization as a function of their tration. At 100 µM MMAF, the tubulin polymerization is inhibited completely. The compounds investigated in the context of the present invention result in a d polymerization rate of the microtubuli. Working Examples 18-21 inhibit the tubulin polymerization at 1 µM to 45-88% of the value measured for 1 µM MMAF.
C-4. In vitro tests for determining cell permeability The cell permeability of a substance can be investigated by means of in vitro testing in a flux assay using Caco-2 cells [M.D. Troutman and D.R. Thakker, Pharm. Res. 20 (8), 1210-1224 (2003)]. For this purpose, the cells were cultured for 15-16 days on 24-well filter plates. For the ination of permeation, the respective working example was applied in a HEPES buffer to the cells either apically (A) or y (B) and incubated for 2 hours. After 0 hours and after 2 hours, s were taken from the cis and trans compartments. The samples were separated by HPLC (Agilent 1200, Böblingen, y) using reverse phase columns. The HPLC system was coupled via a Turbo Ion Spray Interface to a Triple Quadropol mass spectrometer API 4000 (Applied Biosystems Applera, Darmstadt, Germany). The bility was evaluated on the basis of a Papp value, which was ated using the formula published by Schwab et al. [D. Schwab et al., J. Med. Chem. 46, 1716-1725 (2003)].
Of critical importance for toxophores which are released intracellularly is the bility from B to A [Papp (B-A)]: the lower this permeability, the longer the nce time of the working example in the cell following intracellular release, and hence also the longer the time available for interaction with the biochemical target (in this case: tubulin).
Table 7 below sets out permeability data for representative working examples from this assay: Table 7 Working example Papp (B-A) [nm/s] 18 2 19 1 21 2 22 2 23 1 36 1.5 37 0.9 The working examples exhibit a low permeability from B to A [Papp (B-A) and therefore have a long residence time in the CaCo-2 cells. In comparison, monomethylauristatin E (MMAE) and monomethylauristatin F (MMAF) in this test t a Papp (B-A) value of 73 nm/s, and therefore have a significantly shorter residence time in the Caco-2 cells.
C-5. In vitro tests for determining the substrate properties for P-glycoprotein (P-gp) Many tumour cells express transporter proteins for drugs, and this frequently accompanies the development of ance towards cytostatics. Substances which are not substrates of such transporter proteins, such as P-glycoprotein (P-gp) or BCRP, for e, could therefore exhibit an improved activity profile.
The substrate properties of a substance for P-gp ) were determined by means of a flux assay using LLC-PK1 cells which overexpress P-gp (L-MDR1 cells) [A.H. Schinkel et al., J. Clin. . 96, 1698-1705 (1995)]. For this purpose, the LLC-PK1 cells or L-MDR1 cells were cultured on 96-well filter plates for 3-4 days. For determination of the permeation, the respective test substance, alone or in the ce of an tor (such as Ivermectin or Verapamil, for example), was applied in a HEPES buffer to the cells either apically (A) or basally (B) and incubated for 2 hours. After 0 hours and after 2 hours, samples were taken from the cis and trans compartments. The samples were separated by HPLC using reverse phase columns. The HPLC system was coupled via a Turbo Ion Spray Interface to a Triple Quadropole mass ometer API 3000 (Applied Biosystems Applera, Darmstadt, Germany). The permeability was evaluated on the basis of a Papp value which was calculated using the formula hed by Schwab et al. [D.
Schwab et al., J. Med. Chem. 46, 1716-1725 (2003)].
Of critical importance for toxophores which are released intracellularly is the permeability from B to A [Papp (B-A)]: the lower this permeability, the longer the residence time of the working example in the cell following intracellular release, and hence also the longer the time available for interaction with the biochemical target (in this case: tubulin).
Table 8 below lists permeability data for representative working examples from this assay, which was carried out in L-MDR1 cells: Table 8 Working example Papp (B-A) [nm/s] 18 6 19 4 21 3 22 4 23 4 The working examples t a low permeability from B to A [Papp (B-A) and therefore have a long residence time in the L-MDR1 cells.
C-6. Pharmacokinetics in the SNU-16 tumour model Following intravenous administration of various ADCs, the plasma concentrations and tumour concentrations of ADC and also of potential metabolites are measured and the cokinetic parameters such as clearance (CL), area under the curve (AUC) and half-life (t1/2 ) are calculated.
Analysis for quantifying the potentially ing metabolites The measurement of the compounds in plasma and tumour takes place following precipitation of the proteins with methanol, by means of high-pressure liquid chromatography (HPLC) coupled to a tandem mass spectrometer (MS).
For the processing of 100 µL of plasma, it is admixed with 400 µL of methanol and 10 µL of internal standard (ISTD, 50 ng/mL in methanol) and shaken for 10 seconds. After centrifuging for minutes at 16 000 g, 250 µL of supernatant are transferred to an autosampler vial, which was made up with 250 µL of ammonium e buffer (AAC, 10 mM, pH 6.8) and shaken again.
For the sing of a tumour, it is admixed with 4 times the amount of methanol. In a Tissuelyser II (Quiagen), the sample is comminuted at 30 impacts per minute for 6 minutes and then centrifuged off at 16 000 g for 5 minutes. 50 µL of the supernatant are erred to an autosampler vial and made up with 50 µL of ammonium acetate buffer (10 mM, pH 6.8) and with µL of ISTD. After again being shaken, the tumour sample is ready for measurement.
The measurement of both matrix samples takes place, lastly, with the aid of an HPLC-coupled, atmospheric pressure ionization/tandem mass spectrometer by means of a Turbo Ion Spray Interface (TISP) on an API4000 instrument from SCIEX.
HPLC/LC-MSMS (TISP) analysis runs on an HP1100 pump (Agilent) with a Gemini column (5 µm C18 110 A, 50 × 3 mm, enex).
C-7. Activity test in vivo The activity of the conjugates of the invention was tested in vivo by means for example of xenograft models. The skilled person knows of methods in the prior art for testing the activity of a ate of the invention (see, for example, ; Polson et al., Cancer Res. 2009 Mar 15;69(6):2358-64). For this purpose, for example, rodents (e.g. mice) were implanted with a tumour cell line which expresses the target molecule of the binder. These -carrying rodents were subsequently administered either a ate of the invention or a control antibody conjugate, or isotonic salt solution. Administration took place singly or more often. The tumour growth was ined twice a week by means of a g caliper. After a tumour growth of several weeks, the tumour size of conjugate-treated animals and the control group was compared.
The conjugate-treated animals showed a significantly lower tumour size.
C-7a. Testing of ADCs in experimental tumours in the mouse Human tumour cells which express FGFR2 were inoculated subcutaneously into the flank of immunosuppressed mice, such as nude mice or SCID mice. 1-10 million cells are detached from the cell culture, centrifuged and resuspended with 100 µl of medium, 50% medium/50% Matrigel or 100% Matrigel. The cell suspension was injected beneath the skin of the mouse.
Within a few days, a tumour grew. Treatment began no r than at a tumour size of 20-25 mm² after establishment of a tumour.
Treatment with conjugate took place via the intravenous route into the caudal vein of the mouse.
The conjugate was dissolved in PBS and is administered with a volume of 5-10 ml/kg.
The treatment scheme was governed by the pharmacokinetics of the antibody. As a standard, treatment took place three times following every fourth day or every seventh day. Treatment, r, may also be continued further, or a second cycle with three days of treatment may follow at a later point in time.
As a standard basis, 8 animals were used per treatment group. This number may be higher if particularly strong fluctuations in tumour growth or after treatment are anticipated. As well as the groups which e the active substances, one group, as a control group, was treated only with the buffer, in accordance with the same .
In the course of the experiment, the area of the tumour was measured regularly using a sliding caliper in two dimensions (length/width). The area of the tumour was calculated by means of the formula length × width.
At the end of the experiment, the tumours were d and d. The ratio of the average tumour weights for the therapy group (T) to the control group (C) was expressed as T/C. If l and treatment groups finished at different times, then the T/C value was ated using the tumour areas of the last common measurement of all treatment and control groups.
C-7b. g of FGFR2-ADC in the SNU-16 xenograft model in the mouse Two million SNU-16 stomach carcinoma cells were inoculated subcutaneously into the flank of female NODscid mice.
Intravenous treatment with the conjugates was commenced at an average tumour size of -30 mm2. When the control groups had reached the maximum p ermissible size, these groups were ended. The mental groups treated with FGFR2 conjugates were ended when the tumours began to grow again. The activity of the conjugates was determined on day 31, the last point in time at which the vehicle control was still in the experiment. All of the FGFR2 conjugates tested inhibited tumour growth in a dose-dependent manner. At a dose of 5 mg/kg, Example 1 ed a T/C of 0.08, Example 3 a T/C of 0.06 and Example 26 a T/C of 0.10. For all of the animals treated, the tumour at this point in time was smaller than at the beginning of treatment (partial regression of the tumour). At a dose of 2.5 mg/kg, Example 26 achieved a T/C of 0.14. At this dose, Example 26 led to partial sion in 40% of the animals. At the dose of 1 mg/kg, Example 1 achieved a T/C of 0.15 and Example 3 of 0.36. At this dose as well, e 1 leads to partial regression in all of the animals treated, whereas in the case of Example 3 no partial regressions were obtained. Significant umour effect in comparison to the control was achieved for all of the conjugates , up to a dose of 1 mg/kg. The corresponding control antibody-conjugates showed no activity at all in this model at the same doses.
C7c g of FGFR2-ADC in the MFM-223 xenograft model in the mouse million MFM-223 breast carcinoma cells were inoculated subcutaneously into the flank of female NMRI nu/nu mice. These mice had been mented beforehand with estradiol pellets.
Intravenous treatment with the conjugates was commenced at an average tumour size of 30-35 mm2. When the control groups had reached the maximum permissible size on day 40, all of the treatment groups were ended and the tumour weights were ascertained. Example 26, with a dose of 10 mg/kg, achieved a T/C of 0.09, a T/C of 0.13 at a dose of 5 mg/kg and a T/C of 0.26 at a dose of 1 mg/kg. Significant anti-tumour effect in comparison to the control was ed for all three doses tested. The corresponding control antibody conjugate showed a significant non- specific effect in this model only at the dose of 10 mg/kg.
C7d Testing of FGFR2-2 ADC in the NCI-H716 xenograft model in the mouse 1.5 million NCI-H716 intestinal carcinoma cells were ated aneously into the flank of female NMRI nu/nu mice.
Intravenous treatment with the conjugates was commenced at an average tumour size of -30 mm2. When the control groups had reached the maximum sible size on day 36, all of the treatment groups were ended and the tumour weights were ascertained. Treatment with Example 26 resulted in a significant ion in tumour weight, achieving a T/C of 0.24 at mg/kg. At this dose, the corresponding control conjugate had no activity at all in this model. 2012/075277 D. Working examples for pharmaceutical compositions The compounds of the ion can be converted as follows into pharmaceutical ations: i.v. Solution: The compound of the invention is dissolved at a concentration below the saturation solubility in a physiologically tolerated solvent (e.g. isotonic saline solution, D-PBS, or a formulation with glycine and sodium chloride in citrate buffer with addition of rbate 80). The solution is subjected to sterile filtration and dispensed into sterile and pyrogen-free injection containers. i.v. Solution: The compounds of the invention can be converted into the administration forms cited. This can be accomplished in a known way by “mixing with” or “dissolving in” inert, non-toxic, pharmaceutically suitable excipients (e.g. buffer substances, stabilizers, solubilizers, preservatives). The following, for example, may be present: amino acids ne, ine, methionine, arginine, , leucine, isoleucine, ine, glutamic acid, phenylalanine and others), sugars and related compounds se, saccharose, mannitol, trehalose, sucrose, mannose, lactose, sorbitol), glycerol, sodium salts, potassium, ammonium salts and calcium salts (e.g. sodium chloride, ium chloride or disodiumhydrogenphosphate and many others), e/acetic acid buffer systems, phosphate buffer systems, citric acid and citrate buffer systems, trometamol (TRIS and TRIS salts), Polysorbates (e.g. Polysorbate 80 and Polysorbate 20), Poloxamers (e.g. Poloxamer 188 and Poloxamer 171), Macrogols (PEG derivatives, e.g. 3350), Triton X-100, EDTA salts, glutathione, albumins (e.g. human), urea, benzyl alcohol, phenol, chlorocresol , metacresol, benzalkonium chloride and many others. lizate for subsequent conversion into an i.v., s.c. or i.m. solution: Alternatively the compounds of the invention may be converted into a stable lyophilizate (possibly with the aid of abovementioned excipients) and, before being administered, reconstituted with a suitable solvent (e.g. injection-grade water, ic saline solution) and administered.

Claims (36)

Claims
1. Binder-drug ates of the general formula (Ia) H C R35 H C 3 CH 3 O 3 H H N N N D AK G L1 B L2 N N O O CH O CH O O CH 3 3 CH 3 H C CH 3 3 3 (Ia), in which n is a number from 1 to 50, AK is a binder which is an antibody or an antigen-binding antibody fragment that binds to FGFR2, the group §-G-L1-B-L2-§§ is a linker, where § marks the linkage site with the group AK and §§ marks the linkage site with the nitrogen atom, G is a carbonyl or a group of the formula O O HO #1 #1 #2 O N H N #2 H #1 N #2 O , OH O or #1 where #1 marks the linkage site with the sulphur atom of the binder, #2 marks the linkage site with the group L1, L1 is a bond, linear (C1-C10)-alkanediyl, or a group of the a ##1 ##2 O m or ##1 L1A B1 L1B ##2 where m is a number from 2 to 6, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, L1A is linear (C2-C10)-alkanediyl, B1 is a group of the formula O O O ##5 ##6 L5 ##6 N N ##5 N N L6 R29 R30 , R31 R32 or S ##6 ##5 S in which ## 5 marks the linkage site with the group L1A, ## 6 marks the linkage site with the group L1B, L5 is a bond or (C2-C4)-alkanediyl, L6 is a bond or a group of the formula R33 HN HN O O ##8 or ##8 ##7 ##7 R34 O in which ## 7 marks the linkage site with the carbonyl group, ## 8 marks the linkage site with L1B, R33 is hydrogen, (C1-C4)-alkylcarbonyl, tert-butyloxycarbonyl, or benzyloxycarbonyl, R34 is en or methyl, R29 is hydrogen or (C1-C4)-alkyl, R30 is hydrogen or (C1-C4)-alkyl, R29 and R30 together with the atoms to which they are bonded form a 5- or ered heterocycle, R31 is hydrogen or (C1-C4)-alkyl, R32 is hydrogen or (C1-C4)-alkyl, R31 and R32 together with the atoms to which they are bonded form a 5- or 6-membered heterocycle, L1B is linear (C2-C10)-alkanediyl, where 0)-alkanediyl is optionally substituted by 1 to 4 substituents selected independently of one another from the group consisting of methyl, hydroxy and benzyl, where two carbon atoms of the alkanediyl chain in 1,2, 1,3 or 1,4-relation to one r, with inclusion of any carbon atoms situated between them, are optionally bridged to form a (C3-C6)-cycloalkyl ring or a phenyl ring, B is a bond or a group of the formula O O O O * ** L3 ** O ** N N * N N L4 * Q1 R14 R15 R16 R17 O , , , R20 O * P ** * N ** Q2 * ** O , R18 R19 O , R21 R22 , O O R36 O ** ** or N * N O ** H * R23 R24 O R27 , R37 O where * marks the linkage site with L1, ** marks the linkage site with L2, P is O or NH, L3 is a bond or (C2-C4)-alkanediyl, L4 is a bond or a group of the formula R28 HN HN O O **** or *** **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is en or methyl, R28 is hydrogen, (C1-C4)-alkylcarbonyl, tert-butyloxycarbonyl, or benzyloxycarbonyl, Q1 is a 4- to 7-membered heterocycle, Q2 is a 3- to 7-membered carbocycle or a 4- to ered heterocycle, R14 is en or (C1-C4)-alkyl, R15 is hydrogen or (C1-C4)-alkyl, R14 and R15 together with the atoms to which they are bonded form a 5- or 6- membered heterocycle, R16 is hydrogen or (C1-C4)-alkyl, R17 is hydrogen or (C1-C4)-alkyl, R16 and R17 together with the atoms to which they are bonded form a 5- or 6- membered heterocycle, R18 is hydrogen or (C1-C4)-alkyl, R19 is hydrogen or the side group of a natural α-amino acid or of its homologues or isomers, R20 is hydrogen or (C1-C4)-alkyl, R19 and R20 together with the atoms to which they are bonded form a pyrrolidinyl ring, R21 is hydrogen or )-alkyl, R22 is hydrogen or (C1-C4)-alkyl, R21 and R22 together with the atoms to which they are bonded form a 3- to 7- membered carbocycle, R23 is (C1-C4)-alkyl, R24 is hydrogen or (C1-C4)-alkyl, R27 is hydrogen or (C1-C4)-alkyl, R36 is en, (C1-C4)-alkylcarbonyl, tert-butyloxycarbonyl, or oxycarbonyl R37 is hydrogen or methyl, R36 and R37 together with the atoms to which they are bonded form a pyrrolidine ring, L2 is linear (C2-C10)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, where (C2-C10)-alkanediyl is optionally substituted by 1 to 4 substituents selected independently of one another from the group consisting of methyl, hydroxyl, and benzyl, where two carbon atoms of the alkanediyl chain in 1,2-, 1,3- or 1,4-relation to one another, with inclusion of any carbon atoms situated between them, are optionally bridged to form a (C3-C6)-cycloalkyl ring or a phenyl ring, D is a group of the formula O T1 #3 R5 N or #3 R3 R4 O , R26 T2 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen or methyl, R2 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, 1- hydroxyethyl, 4-hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxy enzyl, ylethyl, diphenylmethyl, 1H-imidazolylmethyl, or 1H-indolylmethyl, R1 and R2 er with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula O O O or O N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy, or benzyloxy, R3 is hydrogen or methyl, R4 is isopropyl, isobutyl, sec-butyl, tert-butyl, phenyl, benzyl, 1- yethyl, 4-hydroxybenzyl, 4-hydroxynitrobenzyl, 4-hydroxy aminobenzyl, ylethyl, diphenylmethyl, 1H-imidazolylmethyl, or olylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the e site with the group T1, T1 is a group of the formula -C(=O)-OR7, -C(=O)-NR8R9, -C(=O)-NH-NHR10 or -CH2-O-R11, in which R7 is en, methyl, ethyl, n-propyl, tert-butyl, benzyl, or adamantylmethyl, R8 is hydrogen or methyl, R9 is hydrogen, methyl, ethyl, n-propyl, or benzyl, R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to ered heterocycle, R10 is benzoyl, R11 is benzyl, which is optionally substituted in the phenyl group by methoxycarbonyl or carboxyl, R5 is hydrogen, methyl or a group of the formula #9 O #9 S , , #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHC(R26)-T2, R12 is phenyl which is optionally substituted by methoxycarbonyl, carboxyl, or a group of the formula OH, R13 is phenyl which is optionally substituted by methoxycarbonyl or carboxyl, R26 is hydrogen or hydroxy, T2 is phenyl, benzyl, 1H-indolyl, or 1H-indolylmethyl, R35 is methyl or hydroxy, and also their salts, solvates and solvates of the salts.
2. Binder-drug conjugates of the general formula (Ia) according to Claim 1, in which AK is AK1 or AK2 where AK1 is a binder which is an dy or an antigen-binding antibody fragment that binds to FGFR2 and is bonded via a r atom of the binder to the group G, AK2 is a binder which is an antibody or an antigen-binding antibody fragment that binds to FGFR2 and is bonded via a nitrogen atom of the binder to the group G, G when AK = AK1, is a group of the formula O O HO #1 #1 #2 O N H N #2 H #1 N #2 O , OH O or #1 when AK = AK2, is carbonyl, and also their salts, solvates and solvates of the salts.
3. Binder-drug conjugates of the general formula (Ia) according to Claim 1 or 2, in which n is a number from 1 to 20, AK is AK1 or AK2 where AK1 is a binder which is an antibody or an antigen-binding antibody fragment that binds to FGFR2 and is bonded via the sulphur atom of a ne residue of the binder to the group G, AK2 is a binder which is an antibody or an antigen-binding antibody nt that binds to FGFR2 and is bonded via the NH side group of a lysine residue of the binder to the group G, G when AK = AK1, is a group of the formula N #2 in which #1 marks the linkage site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, when AK = AK2, is carbonyl, L1 is a bond, linear (C2-C6)-alkanediyl, or a group of the formula ##1 ##2 O m or ##1 L1A B1 L1B ##2 where m is a number from 2 to 6, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, L1A is linear )-alkanediyl, B1 is a group of the formula O O O ##5 ##6 L5 ##6 N N ##5 N N L6 R29 R30 , R31 R32 or S ##6 ##5 S in which ## 5 marks the linkage site with the group L1A, ## 6 marks the linkage site with the group L1B, L5 is a bond, L6 is a bond or a group of the formula R33 HN HN O O ##8 or ##8 ##7 ##7 R34 O in which ## 7 marks the linkage site with the carbonyl group, ## 8 marks the linkage site with L1B, R33 is hydrogen, methylcarbonyl, or tert-butyloxycarbonyl, R34 is en or methyl, R29 is hydrogen, R30 is hydrogen, R31 is hydrogen or methyl, R32 is hydrogen or methyl, L1B is linear (C2-C6)-alkanediyl, where (C2-C6)-alkanediyl is optionally substituted by 1 or 2 methyl tuents, B is a bond or a group of the formula O O O O * ** L3 ** O ** N N * N N L4 * Q1 R14 R15 R16 R17 O , , , R20 O O * N ** ** O ** N * N H H R18 R19 O , R21 R22 , R23 R24 O O R36 ** or * N O ** R37 O where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a bond or a group of the formula R28 HN HN O O **** or *** **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is hydrogen or methyl, R28 is en, methylcarbonyl, or tert-butyloxycarbonyl, Q1 is a 4- to 7-membered heterocycle, R14 is hydrogen, R15 is hydrogen, R16 is hydrogen or methyl, R17 is hydrogen or methyl, R16 and R17 together with the atoms to which they are bonded form a piperazinyl ring, R18 is hydrogen, R19 is hydrogen, methyl, propanyl, ylpropanyl, or 1- methylpropanyl, R20 is hydrogen or methyl, R19 and R20 together with the atoms to which they are bonded form a pyrrolidinyl ring, R21 is hydrogen or methyl, R22 is hydrogen or methyl, R21 and R22 together with the atoms to which they are bonded form a cyclopropyl ring, R23 is methyl, R24 is hydrogen or methyl, R27 is hydrogen, R36 is hydrogen, methylcarbonyl, or tert-butyloxycarbonyl, R37 is en or methyl, R36 and R37 together with the atoms to which they are bonded form a pyrrolidine ring, L2 is linear (C2-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, where (C2-C10)-alkanediyl is optionally substituted by 1 or 2 methyl substituents, D is a group of the formula O #3 T1 #3 R5 N or #3 R3 R4 O , R26 T2 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is en, R2 is 1-hydroxyethyl, benzyl, 4-hydroxybenzyl, ylethyl, or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula O O O or O N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy, or benzyloxy, R3 is hydrogen, R4 is 1-hydroxyethyl, benzyl, 4-hydroxybenzyl, 1-phenylethyl, or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the nt en atom, #8 marks the linkage site with the group T1, T1 is a group of the a -C(=O)-OR7, -C(=O)-NR8R9, -C(=O)-NH-NHR10 , or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl, or adamantylmethyl, R8 is en or methyl, R9 is hydrogen, methyl, ethyl, n-propyl or benzyl, R8 and R9 together with the nitrogen atom to which they are bonded form a 4- to 7-membered heterocycle, R10 is l, R11 is benzyl, which is optionally substituted in the phenyl group by methoxycarbonyl or carboxyl, R5 is hydrogen, methyl, or a group of the formula #9 O #9 S , , #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHC(R26)-T2, R12 is phenyl which is optionally substituted by methoxycarbonyl, carboxyl, or a group of the formula –S(O)2OH, R13 is phenyl which is optionally substituted by methoxycarbonyl or carboxyl, R26 is hydrogen or hydroxy, T2 is phenyl, benzyl, 1H-indolyl, or 1H-indolylmethyl, and also their salts, es and solvates of the salts.
4. Binder-drug conjugates of the general formula (Ia) ing to any one of Claims 1 to 3, in which n is a number from 1 to 10, AK is AK1 or AK2 where AK1 is a binder which is an antibody or an antigen-binding antibody fragment that binds FGFR2 and is bonded via the sulphur atom of a cysteine residue of the binder to the group G, AK2 is a binder which is an antibody or an antigen-binding antibody nt that binds FGFR2 and is bonded via the NH side group of a lysine residue of the binder to the group G, G when AK = AK1, is a group of the formula N #2 in which #1 marks the linkage site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, when AK = AK2, is carbonyl, L1 is a bond, linear (C2-C6)-alkanediyl, or a group of the formula ##1 ##2 O m where m is a number 2 or 3, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C2-C6)-alkanediyl is optionally substituted by 1 or 2 methyl tuents, B is a bond or a group of the formula O O O L3 ** O ** * N N L4 * Q1 R16 R17 O , , O O or O * ** ** N * N H H R21 R22 R23 R24 O where * marks the linkage site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a bond or a group of the a R28 HN HN O O **** or *** **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is methyl, R28 is hydrogen, methylcarbonyl, or tert-butyloxycarbonyl, Q1 is piperidine-1,4-diyl, R16 is hydrogen or methyl, R17 is hydrogen or methyl, R16 and R17 together with the atoms to which they are bonded form a piperazinyl ring, R21 is hydrogen or methyl, R22 is en or methyl, R21 and R22 together with the atoms to which they are bonded form a cyclopropyl ring, R23 is methyl, R24 is hydrogen, L2 is linear (C2-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number from 2 to 6, ## 3 marks the linkage site with the group B, ##4 marks the linkage site with the nitrogen atom, D is a group of the formula O T1 #3 R5 N or #3 R3 R4 O , R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is 1-hydroxyethyl, benzyl, oxybenzyl, 1-phenylethyl, or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally tuted heterocycle of the a or O O O O N N N N #6 , #6 , #6 #6 in which #6 marks the linkage site with the carbonyl group, R6 is hydrogen, hydroxy, or benzyloxy, R3 is hydrogen, R4 is benzyl, 1-hydroxybenzyl, 1-phenylethyl, or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7, -NR8R9, or -CH2-O-R11, in which R7 is hydrogen, methyl, ethyl, n-propyl, tert-butyl, benzyl, or adamantylmethyl, R8 is en or methyl, R9 is en, methyl, ethyl, n-propyl, or benzyl, R11 is benzyl, which is optionally substituted in the phenyl group by methoxycarbonyl or carboxyl, R5 is hydrogen, methyl or a group of the formula #9 R13 #9 #9 or R12 R12 , N N in which #9 marks the linkage site with –CHCH2phenyl, R12 is phenyl which is optionally substituted by methoxycarbonyl, carboxyl or a group of the formula –S(O)2OH, R13 is phenyl which is optionally substituted by methoxycarbonyl or carboxyl, and also their salts, solvates and solvates of the salts.
5. Binder-drug conjugates of the general formula (Ia) according to any one of Claims 1 to 4, in which n is a number from 1 to 10, AK is AK2 where AK2 is a binder which is an antibody or an antigen-binding antibody fragment that binds FGFR2 and is bonded via the NH side group of a lysine residue of the binder to the group G, G is carbonyl, L1 is a bond, B is a bond, L2 is linear (C3-C6)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ## 3 marks the linkage site with the group B, ##4 marks the e site with the nitrogen atom, D is a group of the a O #3 T1 N or #3 R3 R4 R1 R2 where #3 marks the e site with the nitrogen atom, R1 is hydrogen, R2 is benzyl or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the adjacent nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula in which #6 marks the linkage site with the carbonyl group, R3 is hydrogen, R4 is benzyl, 4-hydroxybenzyl, or 1H-indolylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the e site with the adjacent nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7 or -C(=O)-NR8R9 in which R7 is hydrogen, methyl, ethyl, n-propyl, utyl, benzyl, or adamantylmethyl, R8 is hydrogen, R9 is hydrogen or benzyl, R35 is methyl, and also their salts, solvates and solvates of the salts.
6. Binder-drug conjugates of the general formula (Ia) ing to any one of Claims 1 to 4, in which n is a number from 1 to 10, AK is AK1 where AK1 is a binder which is an antibody or an antigen-binding antibody nt that binds FGFR2 and is bonded via the sulphur atom of a cysteine residue of the binder to the group G, G is a group of the formula N #2 where #1 marks the linkage site with the cysteine residue of the binder, #2 marks the linkage site with the group L1, L1 is a bond, linear (C3-C5)-alkanediyl, or a group of the formula ##1 ##2 O m where m is a number 2 or 3, ## 1 marks the linkage site with the group G, ##2 marks the linkage site with the group B, where (C3-C5)-alkanediyl is optionally substituted by 1 or 2 methyl substituents, B is a bond or a group of the formula O O L3 ** * N N L4 R16 R17 where * marks the e site with L1, ** marks the linkage site with L2, L3 is a bond or ethane-1,2-diyl, L4 is a bond or a group of the formula O **** R25 O in which *** marks the linkage site with the carbonyl group, **** marks the linkage site with L2, R25 is methyl, R28 is hydrogen, methylcarbonyl, or tert-butyloxycarbonyl, R16 is hydrogen or methyl, R17 is hydrogen or methyl, R16 and R17 together with the atoms to which they are bonded form a piperazinyl ring, L2 is linear (C3-C5)-alkanediyl or is a group of the formula ##3 ##4 O p where p is a number 2 or 3, ## 3 marks the linkage site with the group B, ##4 marks the e site with the nitrogen atom, D is a group of the formula O #3 T1 N or #3 R3 R4 R1 R2 where #3 marks the linkage site with the nitrogen atom, R1 is hydrogen, R2 is benzyl or 1H-indolylmethyl, R1 and R2 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #4 #5 in which #4 marks the linkage site with the nt nitrogen atom, #5 marks the linkage site with the carbonyl group, the ring A with the N-O moiety present therein is a mono- or bicyclic, optionally substituted heterocycle of the formula in which #6 marks the linkage site with the carbonyl group, R3 is hydrogen, R4 is benzyl, 4-hydroxybenzyl or olylmethyl, R3 and R4 together with the carbon atom to which they are bonded form a (1S,2R)phenylcyclopropane-1,1-diyl group of the formula #7 #8 in which #7 marks the linkage site with the nt nitrogen atom, #8 marks the linkage site with the group T1, T1 is a group of the formula -C(=O)-OR7 or -C(=O)-NR8R9, in which R7 is hydrogen, methyl, ethyl, n-propyl, utyl, benzyl, or adamantylmethyl, R8 is hydrogen, R9 is hydrogen or benzyl, R35 is methyl, and also their salts, solvates and solvates of the salts.
7. Binder-drug conjugates of the general formula (Ia) according to claim 1, in which D is a group of the following formula, where * is the linkage site to the nitrogen atom and also their salts, es and solvates of the salts.
8. Compounds of the following formula H C CH H C 3 3 3 CH N O 3 H H N AK N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 where AK is an antibody or an antigen-binding antibody fragment that binds FGFR2, and n is a number from 1 to 10, and also their salts, solvates and solvates of the salts.
9. Compounds of the following formula H C CH H C 3 3 3 CH O 3 N H N AK N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 where AK is an antibody or an antigen-binding antibody fragment which binds FGFR2, and n is a number from 1 to 10, and also their salts, solvates and solvates of the salts.
10. Compound of the ing formula H C CH H C 3 3 3 CH O 3 N H N AK2A N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 where AK2A is M048-D01-hIgG1 and n is a number from 1 to 10, and also their salts, solvates and solvates of the salts.
11. Compound of the following formula H C CH H C 3 3 3 CH N O 3 H H N AK2B N N O N N O O O CH O CH O O 3 3 CH H C CH CH 3 3 3 3 where AK2B is M048-D01-hIgG1-b and n is a number from 1 to 10, and also their salts, solvates and solvates of the salts.
12. Process for preparing the compounds according to any one of Claims 1 to 11, of the general formula (Ia), characterized in that a solution of the binder in a buffer [A] is admixed with a suitable reducing agent ed from the group consisting of dithiothreitol or tris(2-carboxyethyl)phosphine hydrochloride, and is subsequently reacted with a compound of the formula (II) O H C R35 H C 3 CH 3 O 3 H H N N N D N L1 B L2 N N O O CH O CH O O 3 3 CH H C CH CH O 3 3 3 (II), in which D, L1, B, L2, and R35 each have the definitions ted in any one of Claims 1 to 11, to give a nd of the formula (I-A) H C R35 H C O 3 CH 3 O 3 H AK1 H N N N D N L1 B L2 N N O O CH O CH O O 3 3 CH CH 3 O H C CH 3 3 3 (I-A), in which n, AK1, D, L1, B, L2 and R35 each have the definitions indicated in any one of Claims 1 to 11, [B] is reacted with a compound of the formula (III) H C R35 H C O 3 CH 3 O 3 O H O H N N N D N L1 B L2 N N O O CH O CH O O 3 CH O 3 3 H C CH CH 3 3 (III), in which D, L1, B, L2 and R35 each have the definitions indicated in any one of Claims 1 to 11, to give a compound of the formula (I-B) H C R35 H C O 3 CH 3 O 3 H AK2 H N N N D L1 B L2 N N O O CH O CH O O 3 CH 3 3 H C CH CH 3 3 (I-B), in which n, AK2, D, L1, B, L2 and R35 each have the definitions ted in any one of Claims 1 to 11.
13. Compounds prepared by the process in Claim 12, where AK1 and AK2 is an antibody which comprises the six CDR sequences of the antibody M048-D01-hIgG1 or M048- D01-hIgG1-b, the variable light and variable heavy chain of the antibody M048-D01- hIgG1 or M048-D01-hIgG1-b or the light and heavy chain of the antibody M048-D01- hIgG1 or M048-D01-hIgG1-b, and also their salts, solvates and solvates of the salts.
14. Binder-drug conjugate according to any one of Claims 1-11, where the binder binds specifically to FGFR2.
15. Binder-drug conjugate according to any one of claims 1-11 and 13-14, where the binder binds to the extracellular N-terminal epitope SLVEDTTLEPE15) of FGFR2.
16. Binder-drug conjugate or compound according to any one of claims 1-11 and 13-15, where the binder, after binding to FGFR2 on the target cell, is alized by the g of the target cell.
17. Binder-drug conjugate according to any one of claims 1-11 and 13-16, where the binder competes in binding to the cancer target molecule FGFR2 with the GAL-FR21, GALFR22 or M048-D01-hIgG1 antibody.
18. Binder-drug conjugate according to any one of claims 1-11 and 13-17, where the binder comprises the amino acid sequence of the CDR sequences of the variable light and heavy chain of the antibody M048-D01-hIgG1 represented in SEQ ID NO:15 (HCDR1 ), SEQ ID NO:16 (H-CDR2), SEQ ID NO:17 (H-CDR3), SEQ ID NO:18 (LCDR1 ), SEQ ID NO:19 (L-CDR2) and SEQ ID NO:20 (L-CDR3), the amino acid sequence of the variable light and heavy chains of the antibody M048- D01-hIgG1, ented in SEQ ID NO:12 (Vl) and SEQ ID NO:11 (Vh), the amino acid sequence of the variable light and heavy chains of the antibody M048- D01-hIgG1-b, represented in SEQ ID NO:14 (Vl) and SEQ ID NO:13 (Vh), the amino acid sequence of the light and heavy chain of the antibody M048-D01- hIgG1-b represented in SEQ ID NO: 9 (light chain) and SEQ ID NO:10 (heavy chain), the amino acid sequence of the light and heavy chain of the antibody M048-D01- hIgG1 represented in SEQ ID NO: 7 (light chain) and SEQ ID NO:8 (heavy chain), or the amino acid sequence of the variable light and heavy chains of the antibody 1 or GAL-FR22.
19. Binder-drug conjugate or compounds according to any one of Claims 1-11 and 13-18 for the ent and/or prophylaxis of illnesses.
20. Binder-drug conjugate or a nd according to any of Claims 1-11 and 13-19 for use in a method for the treatment and/or prophylaxis of hyperproliferative and/or angiogenic diseases.
21. Binder-drug conjugate according to any of Claims 1-11 and 13-20 for producing a medicament for the treatment and/or prophylaxis of hyperproliferative and/or angiogenic diseases.
22. Medicament comprising a binder-drug conjugate or a compound according to any of Claims 1-11 and 13-21, in combination with an inert, non-toxic, pharmaceutically suitable excipient.
23. Medicament comprising a binder-drug conjugate or a nd according to any of Claims 1-11 and 13-21, in combination with one or more yperproliferative, cytostatic or cytotoxic substances.
24. ment according to Claim 22 or 23 for the treatment and/or prophylaxis of hyperproliferative and/or angiogenic diseases.
25. Use of at least one binder-drug conjugate or a compound according to any of Claims 1- 11 and 13-21 in the manufacture of a medicament for the treatment and/or prophylaxis of hyperproliferative and/or angiogenic es in humans and animals.
26. A binder-drug conjugate according to claim 1 substantially as herein described or exemplified.
27. A binder-drug conjugate according to claim 7 substantially as herein described or exemplified.
28. A compound according to claim 8 substantially as herein described or exemplified.
29. A compound according to claim 9 ntially as herein described or exemplified.
30. A nd according to claim 10 ntially as herein described or exemplified.
31. A compound according to claim 11 substantially as herein described or exemplified.
32. A process according to claim 12 substantially as herein described or exemplified.
33. A compound according to claim 13 substantially as herein described or exemplified.
34. A medicament according to claim 22 substantially as herein described or exemplified.
35. A ment according to claim 23 substantially as herein described or exemplified.
36. A use according to claim 25 substantially as herein described or exemplified. WO 87716 -
NZ625745A 2011-12-14 2012-12-12 FGFR antibody drug conjugates (ADCs) and the use thereof NZ625745B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP11193617.5 2011-12-14
EP11193617 2011-12-14
EP12189467.9 2012-10-22
EP12189467 2012-10-22
PCT/EP2012/075277 WO2013087716A2 (en) 2011-12-14 2012-12-12 New antibody drug conjugates (adcs) and the use thereof

Publications (2)

Publication Number Publication Date
NZ625745A NZ625745A (en) 2016-07-29
NZ625745B2 true NZ625745B2 (en) 2016-11-01

Family

ID=

Similar Documents

Publication Publication Date Title
AU2017203928B2 (en) Novel binder-drug conjugates (ADCs) and their use
US20150023989A1 (en) New antibody drug conjugates (adcs) and the use thereof
US10744205B2 (en) Antibody drug conjugates of kinesin spindel protein (KSP) inhibitors with anti-CD123-antibodies
US20180318438A1 (en) Antibody drug conjugates of kinesin spindel protein (ksp) inhibitors with anti-tweakr-antibodies
NZ625745B2 (en) FGFR antibody drug conjugates (ADCs) and the use thereof
NZ615839B2 (en) Novel binder-drug conjugates (adcs) and their use
TW201339175A (en) New binder-drug conjugates (ADCs) and use thereof
TW201302799A (en) New binder-drug conjugates (ADCs) and use thereof