NZ788703A - Glucocorticoid receptor agonist and immunoconjugates thereof - Google Patents

Glucocorticoid receptor agonist and immunoconjugates thereof

Info

Publication number
NZ788703A
NZ788703A NZ788703A NZ78870317A NZ788703A NZ 788703 A NZ788703 A NZ 788703A NZ 788703 A NZ788703 A NZ 788703A NZ 78870317 A NZ78870317 A NZ 78870317A NZ 788703 A NZ788703 A NZ 788703A
Authority
NZ
New Zealand
Prior art keywords
group
alkyl
hydrogen
compound
formula
Prior art date
Application number
NZ788703A
Inventor
Christian Goess
Martin E Hayes
Jr Axel Hernandez
Adrian D Hobson
Christopher C Marvin
Michael J Mcpherson
Jason Z Oh
John T Randolph
Diana Schmidt
Wendy Waegell
Original Assignee
Abbvie 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 Abbvie Inc filed Critical Abbvie Inc
Publication of NZ788703A publication Critical patent/NZ788703A/en

Links

Abstract

Provided herein are glucocorticoid receptor agonist immunoconjugates, glucocorticoid receptor agonists, and methods of using the same, e.g., to treat autoimmune or inflammatory diseases.

Description

- I - GLUCOCORTICOID RECEPTOR AGONIST AND IMMUNOCONJUGATES THEREOF Related Applications This application is a divisional of New Zealand Patent Application No. 748645 (the national phase application of 2017/035518) and claims ty to U.S. ional Application No. 62/344,948, fi led June 2, 2016, and U.S. Provisional Application No. 62/371,134, fi led August 4, 2016, each of which is herein incorporated by refer ence in its entirety.
Field of the Invention The field of the invention generally relates to glucocorticoid receptor agonist immunoconjugates, and methods of making and using the same, e.g., to treat autoimmune or inflammatory diseases.
Background of the Invention Tumor Necrosis Factor alpha (TNFa) plays a central role in the pathophysiology of several human disorders, and anti-TNFa agents (e.g., adalimumab, etanercept, and infliximab) have clinically validated therapeutic y in the treatment of autoimmune and inflammatory ers, such as rheumatoid arthritis, psoriasis and inflammatory bowel disease. Despite their success in the clinic, anti-TNFa biologics are still limited in the maximal efficacy they can achieve in patients, necessitating the identification and development of more potent and effective therapeutics. Patients treated with anti-TNFa biologics may also develop an immunogenic se to the eutic thus limiting its effectiveness.
Therefore anti-TNFa therapies with lower immunogenicity and high efficacy would be useful for further controlling disease.
Synthetic glucocorticoid receptor agonists (e.g., dexamethasone, prednisolone, and budesonide) are a potent class of small molecules used in the treatment of inflammatory disorders, but their utility in the chronic treatment of disease is limited due to severe side effects. Several approaches to retain the anti-inflammatory efficacy of synthetic glucocorticoids while sparing the unwanted toxicities have been described (Rosen, J and Miner, JN ine Reviews 26: 452-64 (2005)). However these methodologies have met with little success. There is a need in the field of autoimmune and inflammatory disease therapeutics to develop eutics with enhanced efficacy and longer on of action compared to anti-TNF antibodies and with l unwanted effects.
BRIEF SUMMARY OF THE ION In one , the present disclosure provides a glucocorticoid receptor agonist immunoconjugate represented by Formulae I-a and 1-b, below, and the pharmaceutically acceptable salts, solvates, or prodrugs thereof. In another aspect, the present disclosure es a glucocorticoid receptor agonist conjugate represented by Formulae I-a and 1-b, below. Glucocorticoid receptor agonist immunoconjugates having ae I-a and 1-b are useful for treating mune diseases such as, but not d to, rheumatoid arthritis, le idiopathic arthritis, psoriatic tis, ankylosing spondylitis, adult Crohn's disease, pediatric Crohn's disease, ulcerative colitis, plaque psoriasis, hidradenitis suppurativa, uveitis, Behcets disease, a spondyloarthropathy, or psoriasis. In one aspect, glucocorticoid receptor agonist immunoconjugates having Formulae I-a and 1-b are useful for treating rheumatoid arthritis. In one , glucocorticoid receptor agonist conjugates having Formulae I-a and 1-b are useful for treating juvenile idiopathic arthritis. In one aspect, glucocorticoid receptor t immunoconjugates having Formulae I-a and 1-b are useful for treating psoriatic arthritis. In one aspect, glucocorticoid receptor agonist immunoconjugates having Formulae I-a and 1-b are useful for treating ankylosing spondylitis. In one , glucocorticoid receptor agonist immunoconjugates having Formulae I-a and 1-b are useful for treating adult Crohn's disease. In one aspect, glucocorticoid receptor agonist immunoconjugates having Formulae I-a and 1-b are useful for treating pediatric Crohn's e.
In one , glucocorticoid receptor agonist immunoconjugates having Formulae I-a and 1-b are useful for treating ulcerative colitis. In one aspect, orticoid receptor agonist immunoconjugates having Formulae I-a and 1-b are useful for treating plaque psoriasis. In one aspect, glucocorticoid receptor agonist immunoconjugates having Formulae I-a and 1-b are useful for treating hidradenitis suppurativa.
In one aspect, glucocorticoid receptor agonist immunoconjugates having Formulae I-a and 1-b are useful for treating uveitis. In one aspect, glucocorticoid receptor agonist immunoconjugates having Formulae I­ a and 1-b are useful for treating Behcets disease. In one aspect, glucocorticoid receptor agonist conjugates having Formulae I-a and 1-b are useful for treating a spondyloarthropathy. In one aspect, glucocorticoid receptor t immunoconjugates having Formulae I-a and 1-b are useful for treating psoriasis.
In another aspect, the present disclosure provides a glucocorticoid receptor agonist represented by Formulae VII, VII-A, VII-B, VIII, VIII-a, , IX, IX-a, and IX-b, or by Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", below, (wherein R7b is hydrogen) and the pharmaceutically acceptable salts, solvates, or prodrugs thereof. In r aspect, the present disclosure provides a glucocorticoid receptor agonist represented by Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or by Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", below, (wherein R7b is hydrogen). nds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', ', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating autoimmune diseases such as, but not limited to, toid arthritis, juvenile idiopathic arthritis, psoriatic arthritis, ankylosing spondylitis, adult Crohn's disease, pediatric Crohn's disease, ulcerative colitis, plaque psoriasis, hidradenitis suppurativa, uveitis, Behcets disease, a spondyloarthropathy, or psoriasis. In one aspect, compounds having Formulae VII, VII-A, VII­ B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', , VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating rheumatoid arthritis. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, , VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX­ a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating juvenile idiopathic arthritis. In one aspect, compounds having ae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', ', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating tic arthritis. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating sing spondylitis. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for ng adult Crohn's disease. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating ric Crohn's disease. In one aspect, compounds having ae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', ', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", ", VIII-b", IX", IX-a", and IX-b", are useful for treating ulcerative colitis. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", , VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating plaque psoriasis. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", , VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating hidradenitis ativa. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, , VIIl-b, IX, IX-a, and IX-b, or Formulae VII', , VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", , VII-B", VIII", VIII-a", ", IX", IX-a", and IX-b", are useful for ng s. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for treating Behcets disease. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", are useful for ng a spondyloarthropathy. In one aspect, compounds having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or Formulae VII', VII-A', VII-B', VIII', VIiia' , VIII-b', IX', IX-a', IX-b', VII", , VII-B", VIII", VIII-a", ", IX", IX-a", and IX-b", are useful for ng psoriasis.
In another aspect, the present disclosure provides compounds represented by Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or by Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", , VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", as synthetic intermediates that can be used to prepare glucocorticoid receptor agonist immunoconjugates having Formulae I-a and 1-b.
In another , the present disclosure provides a pharmaceutical composition comprising a glucocorticoid receptor agonist immunoconjugate represented by Formulae I-a and 1-b, or a orticoid receptor agonist represented by Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or by ae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", and an excipient and/or a pharmaceutically acceptable carrier.
In another aspect, the present disclosure provides a glucocorticoid or agonist immunoconjugate represented by Formulae I-a and 1-b, or a glucocorticoid receptor t represented by Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b or by Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", , VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", for use in treatment ofautoimmune diseases.
In another aspect, the present disclosure es a use of a glucocorticoid receptor agonist immunoconjugates represented by Formulae I-a and 1-b, or a glucocorticoid receptor agonist represented by Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, and IX-b, or by Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", and IX-b", for the manufacture of a medicament for ng autoimmune diseases.
In another aspect, the present disclosure provides methods of ing glucocorticoid receptor agonist immunoconjugates represented by Formulae I-a and 1-b.
BRIEF DESCRIPTION OF THE DRAWINGS/FIGURES Figure 1 shows the proteolytic stability of an ADC ning a steroid and an ADC containing MMAE (monomethyl auristatin E). (See e 76.) Figure 2 shows the kinetics of drug linker loss of steroid ADC in mice. (See Example 77.) Figure 3 shows the activity of a single therapeutic dose response ofanti-mTNFa steroid ADC in a mouse model ofarthritis. (See Example 85.) Figure 4 shows the activity of anti-human TNFa steroid in huTNFa Tg CAIA mouse model of arthritis. (See Example 87.) Figure 5 is a HIC togram showing a heterogenous mixture containing antibodies having zero - molecules attached ("EO" peak), two - molecules attached ("E2" peak), four SM-L-Q- molecules attached ("E4" peak), SM-L-Q- moieties attached ("E6" peak), and eight SM-L-Q­ molecules attached ("E8" peak), depending upon the number of interchain ide bonds that have been reduced. (SM is a radical of a glucocorticosteroid; L is a linker, and Q is a heterobifunctional group or heterotrifunctional group; or Q is absent.) (See Example 74.) Figure 6 is a SEC chromatogram of adalimumab conjugated with a glucocorticosteroid. (See Example 74.) Figure 7 is a line graph showing raw MS data of adalimumab conjugated with a glucocorticosteroid. (See Example 74.) Figure 8 is a line graph showing deconvoluted MS data of umab conjugated with a glucocorticosteroid. Black square and circle represent the ADC with succinimide yzed and unhydrolyzed, respectively. The relative abundance of hydrolyzed and unhydrolyzed ADC is used to determine hydrolysis conversion. (See Example 74.) Figure 9 shows that an anti-TNF d ADC is significantly more effective in reducing ear inflammation in mice than the concurrent combination of the anti-TNF antibody and the steroid or than the anti-TNF antibody alone. (See Example 84.) Figure 10 shows that a single dose of an NF steroid ADC is as effective in reducing paw swelling as 21 days of daily dosing of a steroid. (See Example 85.) Figure 11 shows the change in weights of animals treated with steroid, an anti-TNF antibody, an anti-TNF ADC, or an isotype ADC. (See Example 85.) Figure 12 shows that a single dose of an anti-TNF steroid ADC can reduce established paw swelling, whereas a single dose ofan anti-TNF antibody had a minimal effect. (See e 88.) Figure 13 shows the effect of treatment with an anti-TNF steroid ADC on tarsal bone loss as ed by Micro-Computed Tomography (µCT). (The individual data points (e.g., circles, squares, or triangles) represent individual animals.) (See Example 88.) Figure 14 shows the effect of treatment with an anti-TNF steroid ADC on inflammation. (The individual data points (e.g., s, squares, or triangles) represent individual animals.) (See Example 88.) Figure 15 shows the effect of treatment with an anti-TNF steroid ADC on pannus ion.
(The individual data points (e.g., circles, squares, or triangles) represent individual animals.) (See Example 88.) Figure 16 shows the effect of treatment with an anti-TNF d ADC on bone erosion. (The individual data points (e.g., circles, s, or triangles) represent individual animals.) (See Example 88.) Figure 17 shows the effect of treatment with an anti-TNF steroid ADC on cartilage damage.
(The individual data points (e.g., circles, squares, or triangles) represent individual animals.) (See Example 88.) Figure 18 shows effect of treatment with an anti-TNF steroid ADC on white blood cells in peripheral blood. (The individual data points (e.g., s, squares, or diamonds) represent dual animals.) (See Example 88.) Figure 19 shows effect of treatment with an anti-TNF steroid ADC on neutrophils in peripheral blood. (The individual data points (e.g., circles, squares, or diamonds) represent dual animals.) (See e 88.) Figure 20 shows effect of treatment with an anti-TNF steroid ADC on lymphocytes in peripheral blood. (The individual data points (e.g., circles, squares, or diamonds) represent individual animals.) (See Example 88.) Figure 21 shows effect of treatment with an anti-TNF steroid ADC on monocytes in peripheral blood. (The individual data points (e.g., circles, squares, or diamonds) represent individual animals.) (See Example 88.) Figure 22 shows effect of treatment with an anti-TNF d ADC on eosinophils m peripheral blood. (See Example 88.) Figure 23 shows effect of treatment with an anti-TNF steroid ADC on basophils in peripheral blood. (See Example 88.) Figure 24 shows the activity of an anti-TNF steroid ADC and an anti-CD163 steroid ADC in mouse collagen-induced arthritis. (See Example 89.) DETAILED DESCRIPTION OF THE INVENTION ed herein are glucocorticoid receptor t immunoconjugates, glucocorticoid receptor agonists, and methods ofmaking and using the same.
I. Definitions To facilitate an tanding of the present disclosure, a number of terms and phrases are defined below.
The term "anti-TNF alpha protein" refers to proteins that are e of (i) binding to TNF alpha and (ii) inhibiting binding of soluble TNF-alpha to cell surface TNF receptors (p55 and/or p75) and/or lysing surface TNF alpha or TNF alpha receptor expressing cells in vitro in the presence of ment. Anti-TNF alpha proteins include, for example, anti-TNF antibodies or antigen-binding nts thereof (e.g., adalimumab or infliximab) as well as e TNF receptors (e.g., etanercept).
As used herein, the terms "antibody" and "antibodies" are terms of art and can be used interchangeably herein and refer to a molecule with an antigen-binding site that ically binds an antigen.
The term "antibody" means an immunoglobulin molecule that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule. As used herein, the term "antibody" encompasses intact polyclonal dies, intact monoclonal antibodies, chimeric antibodies, humanized antibodies, human antibodies, fusion proteins comprising an antibody, and any other modified immunoglobulin molecule so long as the dies exhibit the desired biological activity. An antibody can be of any the five major classes of globulins: IgA, IgD, IgE, IgG, and IgM, or subclasses (isotypes) thereof (e.g. IgG 1, IgG2, IgG3, IgG4, IgAl and IgA2), based on the identity of their heavy-chain nt domains referred to as alpha, delta, epsilon, gamma, and mu, respectively. The different classes of globulins have different and well known subunit structures and dimensional configurations. Antibodies can be naked or conjugated to other molecules such as toxins, radioisotopes, etc. As used herein, the term "antibody" encompasses bispecific and multispecific antibodies.
The term "antibody fragment" refers to a n of an intact dy. An "antigen-binding fragment" refers to a portion of an intact dy that binds to an antigen. An antigen-binding fragment can contain the antigenic determining variable regions of an intact antibody. Examples of dy fragments include, but are not limited to Fab, Fab', 2, and Fv fr ag ments, linear antibodies, and single chain antibodies. An "antigen-binding fragment" can be a bispecific or multispecific antigen-binding fragment.
A "blocking" antibody or an "antagonist" antibody is one which inhibits or s biological activity of the n it binds, such as TNF-alpha. In some embodiments, blocking antibodies or antagonist antibodies substantially or completely inhibit the biological ty of the antigen. The biological activity can be reduced by 10%, 20%, 30%, 50%, 70%, 80%, 90%, 95%, or even 100%.
The term "anti-TNF-alpha antibody" or "an antibody that binds to TNF-alpha" refers to an antibody that is capable of binding TNF-alpha with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting TNF-alpha. The extent of binding of an NF-alpha dy to an unrelated, non-TNF-alpha protein can be less than about 10% of the binding of the antibody to TNF-alpha as ed, e.g., by a radioimmunoassay (RIA). In certain embodiments, an antibody that binds to TNF-alpha has a dissociation constant (Kd) of�l µM,�100 nM,�10 nM,�l nM, or�0.l nM.
A "monoclonal" antibody or antigen-binding fragment thereof refers to a homogeneous antibody or antigen-binding fragment population involved in the highly specific recognition and binding of a single antigenic determinant, or epitope. This is in contrast to polyclonal antibodies that typically include different antibodies directed against different antigenic determinants. The term "monoclonal" antibody or antigen-binding fragment thereof encompasses both intact and full-length monoclonal antibodies as well as antibody fragments (such as Fab, Fab', 2, Fv), single chain (scFv) mutants, fusion proteins sing an antibody portion, and any other modified immunoglobulin molecule comprising an antigen recognition site. rmore, lonal" antibody or antigen-binding fragment thereof refers to such antibodies and antigen-binding fragments thereof made in any number of manners including but not limited to by hybridoma, phage ion, recombinant expression, and transgenic animals.
The term "humanized" dy or antigen-binding nt thereof refers to forms of an (e.g. ) antibodies or antigen-binding fragments that are specific immunoglobulin chains, chimeric immunoglobulins, or fragments thereof that contain minimal non-human (e.g., murine) sequences. Typically, humanized antibodies or antigen-binding fragments thereof are human immunoglobulins in which residues from the mentary determining region (CDR) are replaced by es from the CDR of a non-human species (e.g. mouse, rat, rabbit, hamster) that have the desired specificity, affinity, and capability ("CDR grafted") (Jones et al., Nature 321:522-525 (1986); Riechmann et al., Nature 332:323-327 (1988); Verhoeyen et al., Science 34-1536 (1988)). In some instances, the Fv framework region (FR) es of a human immunoglobulin are replaced with the corresponding residues in an antibody or fragment from a non-human species that has the desired specificity, affinity, and capability. The humanized antibody or n-binding nt thereof can be further modified by the substitution of additional residues either in the Fv framework region and/or within the replaced nonhuman residues to refine and optimize antibody or antigen-binding fragment thereof specificity, affinity, and/or capability. In general, the humanized antibody or n-binding fragment thereof will comprise substantially all of at least one, and typically two or three, variable domains containing all or substantially all of the CDR regions that correspond to the man immunoglobulin whereas all or substantially all of the FR regions are those of a human immunoglobulin sus sequence. The zed antibody or antigen-binding fragment thereof can also se at least a portion of an immunoglobulin constant region or domain (F c ), typically that of a human immunoglobulin. Examples of methods used to generate humanized antibodies are described in U.S. Pat. 5,225,539; Roguska et al., Proc. Natl. Acad. Sci., USA, 91(3):969-973 (1994), and Roguska et al., Protein Eng. 9(10):895-904 (1996). In some embodiments, a "humanized antibody" is a resurfaced antibody.
A "variable region" of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination. The variable regions of the heavy and light chain each consist of four framework regions (FR) connected by three complementarity determining regions (CDRs) also known as hypervariable regions. The CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies. There are at least two techniques for ining CDRs: (1) an approach based on cross-species sequence variability (i.e., Kabat et al.
Sequences ofProteins of logical Interest, (5th ed., 1991, National Institutes of Health, Bethesda Md.)); and (2) an approach based on crystallographic studies ofantigen-antibody complexes (Al-lazikani et al (1997) J. Malec. Biol. 273:927-948)). In addition, combinations of these two ches are sometimes used in the art to determine CDRs.
The Kabat numbering system is generally used when referring to a residue in the le domain (approximately residues 1-107 of the light chain and residues 1-113 of the heavy chain) (e.g., Kabat et al., Sequences of Immunological Interest. 5th Ed. Public Health Service, National Institutes of , da, Md. (1991)). Unless explicitly indicated otherwise, the numbering system used herein is the Kabat numbering system.
The amino acid position numbering as in Kabat, refers to the numbering system used for heavy chain variable domains or light chain variable domains of the compilation ofantibodies in Kabat et al., Sequences ofProteins oflmmunological Interest, 5th Ed. Public Health Service, al Institutes of Health, Bethesda, Md. (1991). Using this numbering system, the actual linear amino acid sequence can contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FR or CDR of the le domain. For example, a heavy chain variable domain can include a single amino acid insert (residue 52a according to Kabat) after residue 52 of H2 and inserted es (e.g. residues 82a, 82b, and 82c, etc. according to Kabat) after heavy chain FR residue 82. The Kabat numbering of residues can be determined for a given antibody by alignment at regions of gy of the sequence of the antibody with a "standard" Kabat numbered sequence. Chothia refers d to the location ofthe structural loops (Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)). The end of the Chothia CDR-H l loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; ifneither 35A nor 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35A and 35B are present, the loop ends at 34). The AbM hypervariable s represent a compromise between the Kabat CDRs and Chothia ural loops, and are used by Oxford Molecular's AbM antibody modeling software.
Loop Kabat AbM Chothia L1 L24-L34 L24-L34 L24-L34 L2 L50-L56 L50-L56 L50-L56 L3 L89-L97 L89-L97 L89-L97 H1 H31-H35B 5B 2..34 (Kabat ing) H1 H31-H35 H26-H35 H26-H32 ia Numbering) H2 H50-H65 H50-H58 H52-H56 H3 H95-H102 H95-H102 02 In certain aspects, the CDRs of an antibody or antigen-binding fragment thereof can be determined according to the Chothia numbering scheme, which refers to the location of immunoglobulin structural loops (see, e.g., a C & Lesk AM, (1987), J Mol Biol 196: 901-917; Al-Lazikani Bet al., (1997) J Mol Biol 273: 927-948; Chothia C et al., (1992) J Mol Biol 227: 799-817; Tramontano A et al., (1990) J Mol Biol 215(1): 175-82; and U.S. Patent No. 7,709,226). Typically, when using the Kabat numbering convention, the Chothia CDR-Hl loop is present at heavy chain amino acids 26 to 32, 33, or 34, the Chothia CDR-H2 loop is present at heavy chain amino acids 52 to 56, and the a CDR-H3 loop is present at heavy chain amino acids 95 to 102, while the Chothia CDR-Ll loop is present at light chain amino acids 24 to 34, the Chothia CDR-L2 loop is present at light chain amino acids 50 to 56, and the Chothia CDR-L3 loop is present at light chain amino acids 89 to 97. The end of the Chothia CDR-Hl loop when numbered using the Kabat numbering convention varies n H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; if neither 35A nor 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35A and 35B are present, the loop ends at 34).
In certain aspects, the CDRs of an antibody or n-binding nt thereof can be determined according to the IMGT numbering system as described in Lefranc M-P, (1999) The Immunologist 7: 132-136 and Lefranc M-P et al., (1999) Nucleic Acids Res 27: 209-212. According to the IMGT numbering scheme, VH-CDRl is at positions 26 to 35, VH-CDR2 is at positions 51 to 57, VHCDR3 is at positions 93 to 102, VL-CDR l is at positions 27 to 32, VL-CDR2 is at positions 50 to 52, and VL-CDR3 is at positions 89 to 97.
In certain s, the CDRs of an dy or antigen-binding fragment thereof can be determined according to MacCallum RM et al., (1996) J Mol Biol 262: 732-745. See also, e.g., Martin A.
"Protein Sequence and Structure Analysis of Antibody Variable Domains," in Antibody Engineering, Kontermann and Dubel, eds., Chapter 31, pp. 422-439, Springer-Verlag, Berlin (2001).
In certain aspects, the CDRs of an antibody or antigen-binding nt thereof can be determined according to the AbM numbering scheme, which refers AbM hypervariable regions which represent a mise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular's AbM antibody modeling software (Oxford Molecular Group, Inc.).
The term "human" antibody means an antibody produced by a human or an antibody having an amino acid sequence corresponding to an antibody produced by a human made using any technique known in the art. This definition of a human antibody includes intact or ength antibodies, fragments f, and/or antibodies comprising at least one human heavy and/or light chain polypeptide such as, for e, an antibody comprising murine light chain and human heavy chain polypeptides.
The term "chimeric" antibodies refers to antibodies wherein the amino acid sequence of the immunoglobulin molecule is d from two or more species. Typically, the variable region of both light and heavy chains ponds to the variable region of antibodies derived from one species of mammals (e.g. mouse, rat, , etc.) with the d specificity, affinity, and capability while the constant regions are homologous to the sequences in antibodies derived from another (usually human) to avoid ing an immune response in that species.
The term "epitope" or "antigenic determinant" are used interchangeably herein and refer to that portion of an antigen capable of being recognized and specifically bound by a particular antibody.
When the antigen is a polypeptide, epitopes can be formed both from contiguous amino acids and noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained upon protein denaturing, whereas epitopes formed by tertiary folding are typically lost upon protein denaturing. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
"Binding affinity" generally refers to the strength of the sum total of alent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
Unless indicated otherwise, as used herein, ng affinity" refers to intrinsic binding affinity which reflects a 1: 1 interaction n members of a g pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). ty can be measured by common s known in the art, including those described herein. Low-affinity antibodies generally bind antigen slowly and tend to dissociate readily, whereas ffinity antibodies generally bind antigen faster and tend to remain bound longer. A variety of methods of measuring binding affinity are known in the art, any of which can be used for purposes of the present disclosure. Specific rative embodiments are described in the following.
"Or better" when used herein to refer to binding affinity refers to a stronger g n a molecule and its binding partner. "Or better" when used herein refers to a stronger binding, ented by a smaller numerical Kd value. For example, an antibody which has an affinity for an antigen of "0.6 nM or better", the antibody's affinity for the antigen is <0.6 nM, i.e. 0.59 nM, 0.58 nM, 0.57 nM etc. or any value less than 0.6 nM.
By "specifically binds," it is generally meant that an antibody binds to an epitope via its antigen binding domain, and that the binding entails some complementarity between the antigen binding domain and the epitope. According to this definition, an antibody is said to "specifically bind" to an e when it binds to that epitope, via its antigen binding domain more readily than it would bind to a , unrelated epitope. The term "specificity" is used herein to qualify the relative affinity by which a certain antibody binds to a certain epitope. For example, antibody "A" may be deemed to have a higher specificity for a given e than antibody "B," or antibody "A" may be said to bind to epitope "C" with a higher specificity than it has for related epitope "D." By "preferentially binds," it is meant that the antibody specifically binds to an epitope more readily than it would bind to a related, similar, homologous, or analogous epitope. Thus, an dy which "preferentially binds" to a given epitope would more likely bind to that epitope than to a related epitope, even though such an antibody may cross-react with the related epitope.
An antibody is said to titively inhibit" binding of a reference antibody to a given epitope if the antibody preferentially binds to that epitope or an overlapping epitope to the extent that it , to some degree, binding of the reference antibody to the epitope. Competitive inhibition may be determined by any method known in the art, for example, competition ELISA assays. An antibody may be said to competitively inhibit binding of the reference antibody to a given epitope by at least 90%, at least 80%, at least 70%, at least 60%, or at least 50%.
The phrase "substantially similar," or "substantially the same", as used herein, denotes a sufficiently high degree of similarity between two numeric values (generally one associated with an antibody of the disclosure and the other associated with a reference/comparator antibody) such that one of skill in the art would consider the difference between the two values to be of little or no ical and/or statistical significance within the context of the biological characteristic measured by said values (e.g., Kd values). The difference between said two values can be less than about 50%, less than about 40%, less than about 30%, less than about 20%, or less than about I0% as a function of the value for the reference/comparator antibody.
A polypeptide, antibody, polynucleotide, , cell, or composition which is "isolated" is a polypeptide, antibody, polynucleotide, vector, cell, or composition which is in a form not found in nature.
Isolated polypeptides, dies, polynucleotides, s, cell or itions include those which have been purified to a degree that they are no longer in a form in which they are found in nature. In some embodiments, an antibody, polynucleotide, vector, cell, or composition which is isolated is substantially pure.
As used herein, "substantially pure" refers to material which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.
The term "immunoconjugate," gate," "antibody-drug conjugate," or "ADC" as used herein refers to a nd or a derivative thereof that is linked to protein such as a cell binding agent (e.g., an anti-TNF-alpha antibody or fragment f) and is defined by a generic formula: (SM-L-Q)n-A, wherein SM = radical d from a small-molecule glucocorticoid receptor agonist, e.g., a glucocorticosteroid, L = linker, Q =heterobifunctional group, a heterotrifunctional group, or is , and A= a protein (e.g., an antibody or antigen-binding fragment thereof, an anti-TNF protein, an NF­ alpha antibody or fragment thereof, a soluble receptor, or a soluble TNF receptor), and n = 1-10.
Immunoconjugates can also be defined by the generic formula in e order: A-(Q-L-SM)n- By way of illustration, the following generic formula shows a immunoconjugate having a dipeptide (Ala-Ala) linker and succinimide her-based heterobifunctional group: L hetrerobifunctional dipeptide group sM '(' NJL,,� N�w-'( s A H l 0 H )-r In the present disclosure, the term "linker" refers to any chemical moiety capable of g a protein, e.g., antibody, antibody fragment (e.g., antigen binding fragments) or functional equivalent to a glucocorticosteroid. Linkers may be susceptible to cleavage (a "cleavable linker") thereby facilitating release of the glucocorticosteroid. For example, such cleavable s may be susceptible to acidinduced cleavage, photo-induced cleavage, peptidase-induced cleavage, esterase-induced cleavage, and disulfide bond cleavage, at conditions under which the glucocorticosteroid and/or the antibody remains active. Alternatively, linkers may be substantially resistant to cleavage (a "noncleavable linker").
In the present disclosure, non-cleavable linkers are any chemical moiety capable of linking a glucocorticosteroid to an antibody in a stable, covalent manner and does not fall off under the categories listed above for cleaveable linkers. Thus, non-cleavable linkers are substantially resistant to acid-induced ge, photo-induced cleavage, peptidase-induced cleavage, esterase-induced cleavage and disulfide bond cleavage. Furthermore, non-cleavable refers to the ability of the chemical bond in the linker or adjoining to the linker to and cleavage induced by an acid, photolabile-cleaving agent, a peptidase, an esterase, or a al or physiological nd that cleaves a disulfide bond, at conditions under which a glucocorticosteroid and/or the antibody does not lose its activity.
Some cleavable linkers are cleaved by peptidases ("peptidase cleavable s"). Only certain peptides are readily cleaved inside or outside cells, see e.g. Trout et al., 79 Proc. Natl. Acad. Sci.
USA, 626-629 (1982) and Umemoto et al. 43 Int. J. Cancer, 677-684 (1989). rmore, es are composed of a-amino acid units and peptidic bonds, which chemically are amide bonds between the carboxylate of one amino acid and the amino group of a second amino acid. Other amide bonds, such as the bond between a carboxylate and the a-amino acid group of lysine, are understood not to be peptidic bonds and are considered non-cleavable.
Some linkers are cleaved by esterases ("esterase cleavable linkers"). Only certain esters can be cleaved by esterases present inside or outside of cells. Esters are formed by the condensation of a carboxylic acid and an alcohol. Simple esters are esters produced with simple alcohols, such as aliphatic alcohols, and small cyclic and small ic alcohols.
In some embodiments, the cleavable linker component may se a peptide comprising one to ten amino acid residues. In these embodiments, the peptide allows for cleavage of the linker by a protease, y facilitating e of the glucocorticosteroid upon exposure to ellular proteases, such as mal enzymes (Doronina et al. (2003) Nat. Biotechnol. -784). Exemplary es include, but are not limited to, ides, tripeptides, tetrapeptides, and pentapeptides. Exemplary dipeptides include, but are not limited to, alanine-alanine (ala-ala), valine-citrulline (vc or t), alanine-phenylalanine (af or ala-phe); phenylalanine-lysine (fk or phe-lys); phenylalanine-homolysine (phe-homolys); and N-methyl-valine-citrulline (Me-val-cit). Exemplary tides include, but are not limited to, glycine-valine-citrulline (gly-val-cit) and glycine-glycine-glycine (gly-gly-gly).
A peptide may comprise naturally-occurring and/or non-natural amino acid residues. The term "naturally-occurring amino acid" refer to Ala, Asp, Cys, Glu, Phe, Gly, His, He, Lys, Leu, Met, Asn, Pro, Gin, Arg, Ser, Tur, Val, Trp, and Tyr. "Non-natural amino acids" (i.e., amino acids do not occur naturally) include, by way of non-limiting example, homoserine, homoarginine, citrulline, phenylglycine, e, iodotyrosine, seleno-cysteine, norleucine ("Nle"), norvaline ("Nva"), beta-alanine, L- or D-naphthalanine, omithine ("Om"), and the like. Peptides can be designed and optimized for enzymatic cleavage by a particular enzyme, for example, a tumor-associated protease, cathepsin B, C and D, or a plasmin protease.
Amino acids also e the D-forms of l and non-natural amino acids. "D-" designates an ammo acid having the "D" (dextrorotary) configuration, as opposed to the configuration in the naturally occurring ("L-") amino acids. Natural and non-natural amino acids can be purchased commercially (Sigma Chemical Co., Advanced Chemtech) or synthesized using methods known in the art.
In the present disclosure, the term "glucocorticosteroid" refers to naturally-occurring or synthetic steroid hormones that interact with glucocorticoid receptors. Non-limiting exemplary glucocorticosteroids include: OH OH 0 0 i= flunisolide triamcinolone acetonide budesonide (F o)y s 0 0 .. ,o .. ,o ,,111 0 0 ciclesonide fluticasone propionate beclomethasone diproprionate By way of example, the A-, B-, C-, and D-rings of the steroid skeleton are marked for budesonide.
Glucocorticosteroids are described in A "radical of a glucocorticosteroid" is derived from the removal of one or more hydrogen atoms from a parent glucocorticosteroid. The removal of hydrogen ) facilitates the attachment of the parent glucocorticosteroid to a linker. In one embodiment, the hydrogen atom is removed from any suitable -NH2 group of the parent glucocorticosteroid. In another embodiment, the hydrogen atom is removed from any suitable -OH group of the parent orticosteroid. In another ment, the hydrogen atom is removed from any suitable a -SH group of the parent orticosteroid. In another embodiment, the hydrogen atom is removed from any suitable -N(H)- group of the parent glucocorticosteroid. In another embodiment, the hydrogen atom is removed from any suitable -CH3, -CHr or -CH= group of the parent glucocorticosteroid. In one embodiment, the "radical of a glucocorticosteroid " is a lent l derived from the removal of one en atom from a parent glucocorticosteroid.
In the present disclosure, the term "heterobifunctional group" or the term "heterotrifunctional group" refers to a chemical moiety that connects a linker and protein, e.g., an antibody. Heterobi- and trifunctional groups are characterized as having ent reactive groups at either end of the chemical moiety. Non-limiting exemplary heterobifunctional groups include: \;:�s-l H00C 0 H '?..
II N s,....-<i. \ .,,/"--.../ 0 0 /--...,_ o s-\ \�� {00 A non-limiting exemplary heterotrifunctional group is: The term "drug dy ratio" or "DAR" refers to the number of SMs (i.e., radical derived from a small-molecule glucocorticoid receptor agonist, e.g., a glucocorticosteroid) linked to A (i.e., a protein, e.g., an antibody or antigen-binding fragment thereof, an anti-TNF protein, an anti-TNF-alpha antibody or fragment thereof, a soluble or, or a e TNF receptor). Thus, in the immunoconjugate having the generic a Q)n-A, the DAR is defined by the variable "n." When referring to a compound having formula (SM-L-Q)n-A representing an individual immunoconjugate, the DAR refers to the number of SMs linked to the individual A (e.g., n is an integer of 1 to 10).
When referring to a compound having formula (SM-L-Q)n-A representing a plurality of immunoconjugates, the DAR refers to the average number of SMs linked to the As (e.g., n is an integer or fraction of 1 to 10). Thus, by way of an example, a compound having formula (SM-L-Q)n-A comprising a first immunoconjugate with3 SM per A and a second immunoconjugate with 4 SM per A would have a DAR (i.e., an "n") of3.5.
The term "subject" refers to any animal (e.g., a mammal), including, but not limited to humans, non-human primates, rodents, and the like, which is to be the recipient of a particular treatment. lly, the terms "subject" and "patient" are used interchangeably herein in nce to a human subject.
The term "pharmaceutical formulation" refers to a preparation which is in such form as to permit the biological activity of the active ingredient to be ive, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
The formulationcan be sterile.
An "effective amount" of an immunoconjugate or glucocorticoid receptor agonist as disclosed herein is an amount sufficient to carry out a specifically stated purpose. An "effective amount" can be determined in on to the stated purpose.
The term peutically effective amount" refers to an amount of an immunoconjugate or glucocorticoid receptor t effective to "treat" a disease or disorder in a subject or mammal. A ylactically effective amount" refers to an amount effective to achieve the desired prophylactic result.
Terms such as "treating" or "treatment" or "to treat" or "alleviating" or "to alleviate" refer to therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder. Thus, those in need of treatment e those already diagnosed with or suspected of having the disorder. Prophylactic or preventative measures refer to measures that prevent and/or slow the development of a targeted pathological condition or disorder. Thus, those in need of prophylactic or preventative measures include those prone to have the disorder and those in whom the disorder is to be prevented.
"Polynucleotide," or "nucleic acid," as used interchangeably herein, refer to polymers of nucleotides of any , and e DNA and RNA. The tides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase. A polynucleotide can comprise modified nucleotides, such as methylated nucleotides and their s. If present, modification to the nucleotide structure can be imparted before or after assembly of the r. The sequence of nucleotides can be interrupted by non-nucleotide components. A cleotide can be further modified after polymerization, such as by conjugation with a labeling component. Other types of modifications e, for example, "caps", substitution of one or more of the naturally occurring tides with an , intemucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, cabamates, etc.) and with d linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, ply-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as well as unmodified forms of the polynucleotide(s). r, any of the hydroxyl groups ordinarily present in the sugars can be replaced, for example, by phosphonate groups, phosphate groups, ted by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or can be conjugated to solid supports. The 5' and 3' terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from I to 20 carbon atoms. Other hydroxyls can also be derivatized to standard ting groups. Polynucleotides can also n analogous forms of ribose or deoxyribose sugars that are lly known in the art, including, for example, ethyl-, 2'-O-allyl, 2'-fluoro- or 2'-azido-ribose, carbocyclic sugar analogs, .alpha.-anomeric , epimeric sugars such as arabinose, xyloses or lyx oses, pyranose sugars, furanose sugars, sedoheptuloses, c analogs and abasic nucleoside analogs such as methyl riboside. One or more phosphodiester linkages can be replaced by alternative linking . These alternative linking groups include, but are not limited to, embodiments n phosphate is replaced by P(O)S ("thioate"), P(S)S ("dithioate"), "(O)NR2 ("amidate"), P(O)R, P(O)OR', CO or CH2 ("formacetal"), in which each R or R' is independently H or substituted or unsubstituted alkyl (1-20 C) optionally containing an ether (--) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
The term "vector" means a uct, which is capable of ring, and optionally expressing, one or more gene(s) or sequence(s) of interest in a host cell. Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, d, cosmid or phage s, DNA or RNA expression vectors associated with cationic condensing agents, DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells, such as producer cells..
The terms "polypeptide," "peptide," and "protein" are used interchangeably herein to refer to polymers of amino acids of any length. The polymer can be linear or branched, it can comprise modified amino acids, and it can be interrupted by non-amino acids. The terms also ass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, ylation, tion, ation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides ning one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. It is understood that, e the polypeptides of this disclosure are based upon antibodies, in certain embodiments, the polypeptides can occur as single chains or associated chains.
The terms "identical" or percent "identity" in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when ed and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The t identity can be measured using ce comparison software or thms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments ofamino acid or nucleotide sequences. One such non-limiting example of a sequence alignment algorithm is the algorithm described in Karlin et al, Proc. Natl. Acad.
Sci., 87:2264-2268 (1990), as modified in Karlin et al., Proc. Natl. Acad. Sci., 90:5873-5877 (1993), and incorporated into the NBLAST and XBLAST programs (Altschul et al., Nucleic Acids Res., 25:3389-3402 (1991)). In certain embodiments, Gapped BLAST can be used as bed in Altschul et al., Nucleic Acids Res. 25:3389-3402 (1997). BLAST-2, WU-BLAST-2 (Altschul et al., Methods in Enzymology, 266:460-480 (1996)), ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or Megalign (DNASTAR) are additional ly ble software programs that can be used to align sequences. In certain embodiments, the percent identity between two nucleotide sequences is determined using the GAP program in GCG software (e.g., using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 90 and a length weight of 1, 2, 3, 4, 5, or 6). In certain alternative ments, the GAP program in the GCG re package, which incorporates the algorithm of Needleman and Wunsch (J Mal. Biol. (48):444-453 (1970)) can be used to determine the t identity n two amino acid sequences (e.g., using either a m 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5). Alternatively, in certain embodiments, the percent identity n nucleotide or amino acid sequences is determined using the algorithm of Myers and Miller (CABIOS, 4:11-17 (1989)). For example, the percent identity can be determined using the ALIGN program on 2.0) and using a PAM120 with residue table, a gap length penalty of 12 and a gap penalty of 4.
Appropriate parameters for maximal alignment by particular alignment software can be determined by one skilled in the art. In certain embodiments, the default parameters of the alignment software are used.
In n embodiments, the percentage identity "X" of a first amino acid sequence to a second ce amino acid is calculated as 100 x (Y /Z), where Y is the number of amino acid es scored as identical matches in the alignment of the first and second sequences (as aligned by visual inspection or a particular sequence alignment program) and Z is the total number of residues in the second ce. If the length of a first sequence is longer than the second sequence, the percent identity of the first sequence to the second sequence will be longer than the percent identity of the second sequence to the first sequence.
As a non-limiting example, whether any particular polynucleotide has a certain percentage sequence identity (e.g., is at least 80% identical, at least 85% identical, at least 90% identical, and in some embodiments, at least 95%, 96%, 97%, 98%, or 99% cal) to a reference sequence can, in certain ments, be determined using the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711).
Bestfit uses the local homology algorithm of Smith and Waterman (Advances in Applied Mathematics 2: 482 489 (1981)) to find the best segment of homology n two sequences. When using Bestfit or any other sequence alignment program to determine whether a particular sequence is, for instance, 95% identical to a reference sequence according to the present disclosure, the parameters are set such that the percentage of identity is calculated over the full length of the reference nucleotide sequence and that gaps in homology of up to 5% of the total number of nucleotides in the reference sequence are d.
In some embodiments, two nucleic acids or polypeptides of the disclosure are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid e identity, when compared and aligned for m correspondence, as measured using a sequence comparison thm or by visual inspection. Identity can exist over a region of the sequences that is at least about 10, about 20, about 40-60 residues in length or any integral value there between, and can be over a longer region than 60-80 residues, for example, at least about 90-100 residues, and in some embodiments, the sequences are substantially identical over the full length of the sequences being compared, such as the coding region of a nucleotide sequence for example.
A "conservative amino acid substitution" is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain. Families of amino acid residues having r side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., e, gine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, praline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, ine). For example, substitution of a phenylalanine for a tyrosine is a conservative substitution. In some ments, vative substitutions in the sequences of the polypeptides and antibodies of the disclosure do not abrogate the binding of the dy containing the amino acid sequence, to the antigen(s), e.g., the TNF-alpha to which the antibody binds. Methods of identifying tide and amino acid conservative tutions which do not eliminate antigen binding are well- known in the art (see, e.g., Brummell et al., Biochem. 32: 1180-1 187 (1993); Kobayashi et al., Protein Eng. 12(10):879-884 (1999); and Burks et al., Proc. Natl. Acad. Sci. USA 2-417 (1997)).
In the present disclosure, the term "halo" as used by itself or as part of another group refers to -Cl, -F, -Br, or -I. In one ment, the halo is -Cl or -F.
In the present disclosure, the term "hydroxy" as used by itself or as part of another group refers to -OH.
In the present disclosure, the term "thiol" or the term "sulfhydryl" as used by itself or as part ofanother group refers to -SH.
In the present disclosure, the term "alkyl" as used by itselfor as part of another group refers to unsubstituted straight- or branched-chain aliphatic hydrocarbons containing from one to twelve carbon atoms, i.e., C1_12 alkyl, or the number of carbon atoms designated, e.g., a C1 alkyl such as methyl, a C2 alkyl such as ethyl, a C3 alkyl such as propyl or isopropyl, a C1_3 alkyl such as methyl, ethyl, propyl, or isopropyl, and so on. In one embodiment, the alkyl is a C1_10 alkyl. In another embodiment, the alkyl is a C1_6 alkyl. In another embodiment, the alkyl is a C1_4 alkyl. In r embodiment, the alkyl is a straight chain C1_10 alkyl. In another embodiment, the alkyl is a branched chain C3_10 alkyl. In another embodiment, the alkyl is a straight chain C1_6 alkyl. In another embodiment, the alkyl is a branched chain C3_6 alkyl. In another embodiment, the alkyl is a straight chain C1_4 alkyl. In another embodiment, the alkyl is a branched chain C3_4 alkyl. In another embodiment, the alkyl is a straight or branched chain C3_4 alkyl. Non-limiting exemplary C1_10 alkyl groups include , ethyl, propyl, isopropyl, butyl, sec-butyl, utyl, iso-butyl, 3-pentyl, hexyl, heptyl, octyl, nonyl, and decyl. Non-limiting exemplary C1_4 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, and iso-butyl.
In the present disclosure, the term "optionally substituted alkyl" as used by itself or as part of another group refers to an alkyl that is either tituted or substituted with one, two, or three substituents independently ed from the group consisting of nitro, hydroxy, cyano, haloalkoxy, aryloxy, alkylthio, sulfonamido, alkylcarbonyl, arylcarbonyl, ulfonyl, arylsulfonyl, carboxy, amido, alkoxycarbonyl, thiol, -N(H)C(=O)NH2, and -N(H)C(=NH)NH2, optionally substituted aryl, and ally substituted heteroaryl. In one embodiment, the optionally tuted alkyl is substituted with two substituents. In another embodiment, the optionally substituted alkyl is substituted with one substituent. In another embodiment, the optionally substituted alkyl is unsubstituted. Non-limiting exemplary substituted alkyl groups include -CH2OH, -CH2SH, -CH2Ph, -CH2(4-OH)Ph, -CH2(imidazolyl), 2CO2H, -CH2CH2SO2CH3, -CH2CH2COPh, and -CH2OC(=O)CH3.
In the t disclosure, the term "cycloalkyl" as used by itself or as part of another group refers to unsubstituted ted or partially unsaturated, e.g., containing one or two double bonds, cyclic tic arbons ning one to three rings having from three to twelve carbon atoms, i.e., C3_12 lkyl, or the number of carbons designated. In one embodiment, the cycloalkyl has two rings.
In another embodiment, the cycloalkyl has one ring. In another embodiment, the cycloalkyl is saturated.
In another embodiment, the lkyl is unsaturated. In another embodiment, the lkyl is a C3_8 cycloalkyl. In another embodiment, the cycloalkyl is a C3_6 cycloalkyl. The term "cycloalkyl" is meant to include groups wherein a ring -CH2-is replaced with a -C(=O)-. Non-limiting exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, ctyl, norbomyl, decalin, adamantyl, cyclohexenyl, cyclopentenyl, and cyclopentanone.
In the present sure, the term "optionally substituted cycloalkyl" as used by itself or as part of another group refers to a cycloalkyl that is either unsubstituted or substituted with one, two, or three substituents independently selected from the group consisting of halo, nitro, cyano, hydroxy, alkylcarbonyloxy, cycloalkylcarbonyloxy, amino, haloalkyl, yalkyl, alkoxy, haloalkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, carboxy, carboxyalkyl, optionally substituted alkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxyalkyl, (amino)alkyl, (carboxamido)alkyl, (heterocyclo)alkyl, and -OC(=O)-amino, The term optionally substituted cycloalkyl includes cycloalkyl groups having a fused optionally substituted aryl, e.g., phenyl, or fused optionally substituted heteroaryl, e.g., pyridyl. An optionally substituted cycloalkyl having a fused optionally substituted ary l or fused optionally substituted heteroary l group may be attached to the remainder of the molecule at any available carbon atom on the cycloalkyl ring. In one embodiment, the optionally substituted cycloalkyl is substituted with two substituents. In another embodiment, the optionally tuted cycloalkyl is substituted with one substituent. In another embodiment, the optionally substituted cycloalkyl is unsubstituted.
In the present disclosure, the term "ary l" as used by itself or as part of r group refers to unsubstituted monocyclic or bicyclic aromatic ring systems having from six to fourteen carbon atoms, i.e., a C6_14 ary l. Non-limiting exemplary ary l groups include phenyl (abbreviated as "Ph"), naphthyl, thry l, anthracyl, indenyl, azulenyl, biphenyl, biphenylenyl, and fluorenyl . In one embodiment, the ary l group is phenyl or naphthyl.
In the present disclosure, the term "optionally substituted ary l" as used herein by itself or as part of another group refers to an ary l that is either unsubstituted or substituted with one to five substituents independently selected from the group consisting of halo, nitro, cyano, hydroxy, thiol, amino, alkylamino, dialkylamino, optionally substituted alkyl, haloalkyl, hydroxyalkyl, alkoxy, koxy, ary loxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, ary lcarbonyl, alkylsulfonyl, haloalkylsulfonyl cycloalkylsulfonyl, (cycloalkyl)alkylsulfonyl, ary nyl, heteroary lsulfonyl, heterocyclosulfonyl, carboxy, carboxyalkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted ary l, optionally tuted heteroary l, ally substituted heterocyclo, alkoxycarbonyl, alkoxyalkyl, (amino)alkyl, ( carboxamido)alkyl, and (heterocyclo)alkyl.
In one embodiment, the optionally substituted ary l is an optionally tuted phenyl. In another embodiment, the optionally tuted phenyl has four substituents. In another embodiment, the optionally substituted phenyl has three substituents. In another embodiment, the optionally substituted phenyl has two tuents. In another embodiment, the optionally substituted phenyl has one tuent. In another ment, the optionally tuted phenyl is tituted. Non-limiting exemplary substituted ary l groups include 2-methylphenyl, 2-methoxyphenyl, 2-fluorophenyl, 2- chlorophenyl, ophenyl, ylphenyl, 3-methoxyphenyl, 3-fluorophenyl, 3-chlorophenyl, 4- methylphenyl, 4-ethylphenyl, oxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 2,6-di-fluorophenyl, 2,6- di-chlorophenyl, 2-methyl, 3-methoxyphenyl, 2-ethyl, 3-methoxyphenyl, 3,4-di-methoxyphenyl, 3,5-difluorophenyl 3,5-di-methylphenyl, 3,5-dimethoxy, 4-methylphenyl, 2-fluorochlorophenyl, 3-chloro fluorophenyl, 4-(pyridinylsulfonyl)phenyl The term optionally substituted ary l includes phenyl groups having a fused optionally tuted cycloalkyl or fused optionally substituted heterocyclo group. An optionally substituted phenyl having a fused optionally substituted cycloalkyl or fused optionally substituted heterocyclo group may be attached to the remainder of the molecule at any available carbon atom on the phenyl ring.
In the present disclosure, the term yl" as used by itself or as part of another group refers to an alkyl containing one, two or three carbon-to-carbon double bonds. In one embodiment, the alkenyl has one carbon-to-carbon double bond. In another embodiment, the alkenyl is a C2_6 alkenyl. In r embodiment, the alkenyl is a C2 4 alkenyl. Non-limiting exemplary alkenyl groups include ethenyl, propenyl, penyl, butenyl, sec-butenyl, pentenyl, and hexenyl.
In the present disclosure, the term "optionally tuted alkenyl" as used herein by itself or as part of another group refers to an alkenyl that is either unsubstituted or substituted with one, two or three substituents independently selected from the group consisting of halo, nitro, cyano, hydroxy, amino, alkylamino, dialkylamino, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, carboxy, carboxyalkyl, optionally substituted alkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, heteroaryl, and optionally substituted heterocyclo.
In the present disclosure, the term "alkynyl" as used by itself or as part of another group refers to an alkyl containing one to three carbon-to-carbon triple bonds. In one ment, the alkynyl has one carbon-to-carbon triple bond. In another embodiment, the alkynyl is a C2_6 l. In another embodiment, the alkynyl is a C2 4 alkynyl. Non-limiting exemplary alkynyl groups e ethynyl, propynyl, l, 2-butynyl, pentynyl, and hexynyl groups.
In the present disclosure, the term "optionally tuted alkynyl" as used herein by itself or as part refers to an alkynyl that is either unsubstituted or substituted with one, two or three substituents independently selected from the group consisting of halo, nitro, cyano, hydroxy, amino, alkylamino, dialkylamino, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyloxy, hio, carboxamido, sulfonamido, arbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, carboxy, carboxyalkyl, optionally tuted alkyl, cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, and heterocyclo.
In the present disclosure, the term "haloalkyl" as used by itself or as part of another group refers to an alkyl substituted by one or more fluorine, chlorine, e and/or iodine atoms. In one embodiment, the alkyl group is substituted by one, two, or three fluorine and/or chlorine atoms. In another embodiment, the haloalkyl group is a C1_4 haloalkyl group. Non-limiting exemplary kyl groups include methyl, 2-fluoroethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, fluoroethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, 3,3,3-trifluoropropyl, trifluorobutyl, and trichloromethyl groups.
In the present sure, the term "alkoxy" as used by itself or as part of another group refers to an optionally substituted alkyl, ally tuted cycloalkyl, optionally substituted alkenyl, or optionally substituted alkynyl attached to a terminal oxygen atom. In one embodiment, the alkoxy is an optionally substituted alkyl attached to a terminal oxygen atom. In one embodiment, the alkoxy group is a C 1_6 alkyl attached to a terminal oxygen atom. In another embodiment, the alkoxy group is a C 1_4 alkyl attached to a terminal oxygen atom. Non-limiting exemplary alkoxy groups e methoxy, ethoxy, and tert-butoxy.
In the present sure, the term "alkylthio" as used by itself or as part of another group refers to an ally substituted alkyl attached to a terminal sulfur atom. In one embodiment, the alkylthio group is a C 1_4 alkylthio group. Non-limiting exemplary alkylthio groups include -SCH3 and -SCH2CH3.
In the present disclosure, the term "haloalkoxy" as used by itself or as part of another group refers to a haloalkyl attached to a terminal oxygen atom. Non-limiting exemplary haloalkoxy groups include methoxy, difluoromethoxy, trifluoromethoxy, and 2,2,2-trifluoroethoxy.
In the present disclosure, the term "heteroaryl" refers to unsubstituted monocyclic and ic ic ring systems having 5 to 14 ring atoms, i.e., a 5- to 14-membered heteroaryl, wherein at least one carbon atom of one of the rings is replaced with a heteroatom independently selected from the group consisting of oxygen, nitrogen and sulfur. In one embodiment, the heteroaryl contains 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of oxygen, nitrogen and sulfur. In one ment, the heteroaryl has three heteroatoms. In another embodiment, the heteroaryl has two heteroatoms. In another embodiment, the heteroaryl has one atom. In another embodiment, the heteroaryl is a 5- to IO-membered heteroaryl. In another embodiment, the heteroaryl is a 5- or 6-membered heteroaryl. In another embodiment, the heteroaryl has 5 ring atoms, e.g., thienyl, a ered heteroaryl having four carbon atoms and one sulfur atom. In another embodiment, the heteroaryl has 6 ring atoms, e.g., pyridyl, a ered heteroaryl having five carbon atoms and one nitrogen atom. Non-limiting exemplary aryl groups include thienyl, benzo[b]thienyl, naphtho[2,3- b]thienyl, thianthrenyl, furyl, benzofuryl, pyranyl, isobenzofuranyl, benzooxazonyl, chromenyl, xanthenyl, 2H-pyrrolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, isoindolyl, 3H-indolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, cinnolinyl, quinazolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, �-carbolinyl, phenanthridinyl, nyl, pyrimidinyl, phenanthrolinyl, phenazinyl, thiazolyl, isothiazolyl, phenothiazolyl, olyl, furazanyl, and phenoxazinyl. In one embodiment, the aryl is ed from the group consisting of thienyl (e.g., thienyl and 3-yl), furyl (e.g., 2-furyl and 3-furyl), pyrrolyl (e.g., lH-pyrrolyl and lH­ pyrrolyl), imidazolyl (e.g., 2H-imidazolyl and 2H-imidazolyl), pyrazolyl (e.g., lH-pyrazolyl, lH-pyrazolyl, and lH-pyrazolyl), pyridyl (e.g., pyridinyl, pyridinyl, and pyridinyl), pyrimidinyl (e.g., pyrimidinyl, pyrimidinyl, and pyrimidinyl), lyl (e.g., thiazolyl, lyl, and thiazolyl), azolyl (e.g., isothiazolyl, isothiazolyl, and isothiazolyl), oxazolyl (e.g., oxazolyl, oxazolyl, and oxazolyl), isoxazolyl (e.g., isoxazolyl, isoxazolyl, and isoxazolyl), and indazolyl (e.g., lH-indazolyl). The term "heteroaryl" is also meant to include possible N-oxides. A non-limiting exemplary N-oxide is pyridyl N-oxide.
In one embodiment, the heteroaryl is a 5- or 6-membered heteroaryl. In one embodiment, the aryl is a 5-membered heteroaryl, i.e., the heteroaryl is a monocyclic aromatic ring system having 5 ring atoms wherein at least one carbon atom of the ring is replaced with a heteroatom independently selected from nitrogen, oxygen, and sulfur. Non-limiting exemplary 5-membered heteroaryl groups include thienyl, furyl, pyrrolyl, oxazolyl, pyrazolyl, imidazolyl, thiazolyl, azolyl, and isoxazolyl. In another ment, the heteroaryl is a 6-membered heteroaryl, e.g., the heteroaryl is a monocyclic ic ring system having 6 ring atoms wherein at least one carbon atom of the ring is ed with a nitrogen atom. Non-limiting exemplary ered heteroaryl groups include pyridyl, pyrazinyl, pyrimidinyl, and pyridazinyl.
In the present disclosure, the term "optionally substituted heteroaryl" as used by itself or as part of another group refers to a heteroaryl that is either unsubstituted or substituted with one two, three, or four substituents, independently selected from the group consisting of halo, nitro, cyano, hydroxy, amino, mino, dialkylamino, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyloxy, hio, carboxamido, sulfonamido, arbonyl, arylcarbonyl, alkylsulfonyl, haloalkylsulfonyl cycloalkylsulfonyl, (cycloalkyl)alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, carboxy, carboxyalkyl, optionally substituted alkyl, optionally tuted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cyclo, alkoxyalkyl, (amino)alkyl, (carboxamido)alkyl, and (heterocyclo)alkyl. In one embodiment, the optionally tuted heteroaryl has one tuent. In another embodiment, the optionally substituted heteroaryl is unsubstituted. Any available carbon or nitrogen atom can be substituted. The term optionally substituted heteroaryl includes heteroaryl groups having a fused optionally substituted cycloalkyl or fused ally substituted heterocyclo group. An optionally substituted aryl having a fused optionally substituted cycloalkyl or fused optionally substituted heterocyclo group may be attached to the remainder of the molecule at any available carbon atom on the heteroaryl ring.
In the present disclosure, the term "heterocyclo" as used by itself or as part of another group refers to unsubstituted saturated and partially unsaturated, e.g., containing one or two double bonds, cyclic groups containing one, two, or three rings having from three to fourteen ring s, i.e., a 3-to 14- membered heterocyclo, wherein at least one carbon atom of one of the rings is replaced with a heteroatom. Each heteroatom is independently selected from the group consisting of oxygen, sulfur, including sulfoxide and sulfone, and/or nitrogen atoms, which can be oxidized or quatemized. The term "heterocyclo" includes groups wherein a ring -CHr is replaced with a -C(=O)-, for e, cyclic ureido groups such as 2-imidazolidinone and cyclic amide groups such as �-lactam, am, 8-lactam, am, and piperazinone. The term "heterocyclo" also includes groups having fused optionally substituted aryl groups, e.g., indolinyl or chromanyl. In one embodiment, the heterocyclo group is a C4_6 heterocyclo, i.e., a 4-, 5- or 6-membered cyclic group, containing one ring and one or two oxygen and/or en atoms. In one ment, the heterocyclo group is a C4_6 heterocyclo containing one ring and one nitrogen atom. The heterocyclo can be optionally linked to the rest of the molecule through any available carbon or nitrogen atom. Non-limiting ary heterocyclo groups include azetidinyl, dioxanyl, tetrahydropyranyl, 2-oxopyrrolidiny1, piperazinone, piperazine-2,6-dione, 2-imidazolidinone, piperidinyl, morpholinyl, piperazinyl, pyrrolidinyl, and indolinyl.
In the t disclosure, the term nally substituted heterocyclo" as used herein by itself or part of another group refers to a heterocyclo that is either unsubstituted or substituted with one, two, three, or four substituents independently selected from the group consisting of halo, nitro, cyano, hydroxy, amino, alkylamino, dialkylamino, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, cycloalkylcarbonyl, alkoxycarbonyl, CF3C( O)-, arylcarbonyl, alkylsulfonyl, arylsulfonyl, carboxy, carboxyalkyl, alkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, ally substituted aryl, optionally substituted heterocyclo, alkoxyalkyl, )alkyl, (carboxamido)alkyl, or ocyclo)alkyl. Substitution may occur on any available carbon or nitrogen atom, or both.
In the present disclosure, the term "amino" as used by itself or as part of another group refers to a radical of the formula -NR22aR22\ wherein R22a and R22b are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, and aralkyl, or R22a and R22b are taken together to form a 3- to 8-membered optionally substituted heterocyclo. Non-limiting exemplary amino groups include -NH2 and -N(H)(CH3).
In the present disclosure, the term "carboxamido" as used by itself or as part of another group refers to a radical of formula -C( O)NR23aR23\ n R23a and R23b are each independently selected from the group consisting of hydrogen, optionally substituted alkyl, hydroxyalkyl, and optionally tuted aryl, optionally substituted heterocyclo, and optionally substituted heteroaryl, or R23a and R23b taken together with the nitrogen to which they are attached form a 3- to ered optionally substituted cyclo group. In one ment, R23a and R23b are each independently hydrogen or optionally substituted alkyl. In one embodiment, R23a and R23b are taken together to taken together with the nitrogen to which they are attached form a 3- to 8-membered optionally substituted heterocyclo group.
Non-limiting exemplary carboxamido groups e -CONH2, -CON(H)CH3, and -CON(CH3)2.
] In the present disclosure, the term "alkoxycarbonyl" as used by itself or as part of another group refers to a carbonyl group, i.e., -C( O)-, substituted with an alkoxy. In one embodiment, the alkoxy is a C 1_4 alkoxy. Non-limiting exemplary alkoxycarbonyl groups include -C( O)OMe, -C( O)OEt, and - C( O)OtBu.
In the present disclosure, the term "carboxy" as used by itself or as part of r group refers to a radical of the formula -C02H.
In the present disclosure, the term "maleimide" as used by itself or as part of another group refers to: In the present disclosure, the term "succinimide" as used as part of a cleavable linker refers to: /tr\0 In the present disclosure, the term "hydrolyzed succinimide" as used as part of a cleavable linker refers to: In the present sure, the term "amide" as used as part of a ble linker refers to: In the present disclosure, the term "thiourea" as used as part of a cleavable linker refers to: In the present disclosure, the term ther" as used as part of a cleavable linker refers to: � -s- � In the present disclosure, the term "oxime" as used as part of a cleavable linker refers to: In the present disclosure, the term "self-immolative group" as used as part of a cleavable linker refers to bifunctional chemical moiety that is capable of ntly linking two spaced chemical moieties into a normally stable tripartite molecule, can release one of the spaced chemical moieties from the tripartite molecule by means of enzymatic cleavage; and following enzymatic ge, can spontaneously cleave from the remainder of the le to release the other of the spaced chemical moieties, e.g., a glucocorticosteroid. In some embodiments, a self-immolative group comprises a p-aminobenzyl unit. In some such embodiments, a p-aminobenzyl alcohol is attached to an amino acid unit via an amide bond, and a carbamate, methylcarbamate, or carbonate is made between the benzyl alcohol and the drug (Hamann et al. (2005) Expert Opin. Tuer. Patents (2005) 15:1087-1103). In some embodiments, the self-immolative group is p-aminobenzyloxycarbonyl (PAB).
In the present disclosure, the term "protecting group" or "PG" refers to group that blocks, i.e., protects, the amine functionality while reactions are carried out on other functional groups or parts of the molecule. Those skilled in the art will be familiar with the ion, attachment, and cleavage of amine protecting groups, and will appreciate that many different protective groups are known in the art, the suitability of one protective group or another being dependent on the particular the synthetic scheme d. Treatises on the subject are available for tation, such as Wuts, P. G. M.; Greene, T. W., e's Protective Groups in Organic Synthesis", 4th Ed., J. Wiley & Sons, NY, 2007. Suitable protecting groups include the carbobenzyloxy (Cbz), tert-butyloxycarbonyl (BOC), 9-fluorenylmethyloxycarbonyl (FMOC), and benzyl (Bn) group. In one embodiment, the protecting group is the BOC group.
The compounds disclosed herein contain asymmetric centers and thus give nse to enantiomers, diastereomers, and other stereoisomeric forms. The present disclosure is meant to encompass the use of all such possible forms, as well as their racemic and resolved forms and mixtures thereof. The individual enantiomers can be ted according to methods known in the art in view of the present disclosure. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that they include both E and Z geometric isomers. All tautomers are also intended to be encompassed by the present disclosure.
The present sure encompasses the ation and use of es of the compounds disclosed . Solvates typically do not significantly alter the physiological activity or toxicity of the compounds, and as such may function as pharmacological equivalents. The term "solvate" as used herein is a combination, al ation and/or salvation of a compound of the present disclosure with a solvent molecule such as, e.g. a ate, monosolvate or hemisolvate, where the ratio of solvent molecule to compound of the t disclosure is about 2:1, about 1: 1 or about 1:2, respectively. This physical association es varying degrees of ionic and covalent bonding, including hydrogen g.
In certain instances, the solvate can be isolated, such as when one or more solvent molecules are incorporated into the l lattice of a lline solid. Thus, te" encompasses both solutionphase and isolatable solvates. Compounds disclosed herein can be present as solvated forms with a pharmaceutically acceptable solvent, such as water, methanol, ethanol, and the like, and it is intended that the disclosure includes both solvated and unsolvated forms of nds disclosed herein. One type of solvate is a hydrate. A "hydrate" relates to a particular subgroup of solvates where the solvent molecule is water. Solvates typically can function as pharmacological lents. Preparation of solvates is known in the art. See, for example, M. Caira et al, J ceut. Sci., 93(3):601-611 (2004), which describes the preparation of solvates of fluconazole with ethyl acetate and with water. Similar preparation of solvates, hemisolvates, hydrates, and the like are described by E.C. van Tonder et al., AAPS Pharm. Sci.
Tech., 5(1):Article 12 (2004), and A.L. Bingham et al., Chem. Commun. 603-604 (2001). A l, non-limiting, process of preparing a solvate would involve dissolving a compound disclosed herein in a d solvent (organic, water, or a e thereof) at atures above 20°C to about 25°C, then g the on at a rate sufficient to form crystals, and isolating the crystals by known s, e.g., filtration. Analytical techniques such as infrared spectroscopy can be used to confirm the presence of the solvent in a crystal of the solvate.
The present disclosure encompasses the preparation and use of salts of the compounds disclosed herein, including non-toxic pharmaceutically acceptable salts. Examples of pharmaceutically acceptable addition salts include nic and organic acid addition salts and basic salts. The pharmaceutically acceptable salts include, but are not limited to, metal salts such as sodium salt, potassium salt, cesium salt and the like; alkaline earth metals such as calcium salt, magnesium salt and the like; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N'-dibenzylethylenediamine salt and the like; inorganic acid salts such as hydrochloride, hydrobromide, phosphate, sulphate and the like; organic acid salts such as citrate, lactate, tartrate, maleate, fumarate, mandelate, acetate, dichloroacetate, trifluoroacetate, oxalate, formate and the like; sulfonates such as methanesulfonate, benzenesulfonate, p-toluenesulfonate and the like; and amino acid salts such as arginate, asparginate, glutamate and the like.
Acid addition salts can be formed by mixing a solution of the particular compound sed with a on of a pharmaceutically able non-toxic acid such as hydrochloric acid, fumaric acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, ic acid, phosphoric acid, oxalic acid, dichloroacetic acid, or the like. Basic salts can be formed by mixing a solution of the compound of the present disclosure with a solution of a pharmaceutically acceptable non-toxic base such as sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate and the like.
As used in the present sure and claims, the singular forms "a," "an," and "the" include plural forms unless the context clearly dictates otherwise.
It is tood that er embodiments are described herein with the language ising," otherwise analogous embodiments described in terms of "consisting of' and/or "consisting essentially of' are also provided.
The term "and/or" as used in a phrase such as "A and/or B" herein is intended to include both "A and B," "A or B," "A," and "B." Likewise, the term "and/or" as used in a phrase such as "A, B, and/or C" is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
II. ns for linkage to glucocorticoid receptor agonists The present disclosure provides agents immunoconjugates containing glucocorticoid receptor agonists linked to proteins, for example, antibodies or antigen-binding fragments thereof and soluble receptor proteins. In some embodiments, the antibody or antigen-binding fragment thereof is human, humanized, chimeric, or murine. In some embodiments, the protein, e.g., antibody, antigen-binding fragment f, or soluble receptor protein, can bind to a target on the e of a cell and become internalized.
The present disclosure also provides immunoconjugates containing glucocorticoid receptor agonists linked to anti-TNF alpha proteins. In certain embodiments, the anti-TNF alpha proteins are antibodies or n-binding fragments f. In certain embodiments, the anti-TNF alpha proteins are antibodies or antigen-binding nts thereof that bind to TNF alpha (e.g., soluble TNF alpha and/or membrane bound TNF . In certain embodiments, the anti-TNF alpha ns are soluble TNF receptor proteins, e.g., soluble TNF or proteins fused to a heavy chain nt domain or fragment thereof such as an Fe. In some embodiments, the anti-TNF alpha protein, e.g., NF antibody, antigen-binding fragment thereof, or soluble TNF receptorcan bind to TNF alpha on the surface of a cell and become internalized. For example, US 2014/0294813, which is herein incorporated by referece in its entirety, discloses anti-TNF proteins that exhibit cellular internalization upon binding to cell surface human TNF.
In certain embodiments, the antibodies or antigen-binding fragments thereof bind to human and/or mouse TNF-alpha. Antibodies and antigen-binding fragments that bind to TNF-alpha are known in the art.
The full-length amino acid sequence for membrane bound human TNF alpha is: ] MSTESMIRDVELAEEALPKKTGGPQGSRRCLFLSLFSFLIVAGATTLFCLLHFGVIGPQ REEFPRDLSLISPLAQAVRSSSRTPSDKPVAHVVANPQAEGQLQWLNRRANALLANGVELRDNQ LVVPSEGLYLIYSQVLFKGQGCPSTHVLLTHTISRIAVSYQTKVNLLSAIKSPCQRETPEGAEAKP WYEPIYLGGVFQLEKGDRLSAEINRPDYLDFAESGQVYFGIIAL (SEQ ID NO:1 ). Soluble human TNF alpha contains amino acids 77-233 of SEQ ID NO:1. The full-length amino acid sequence for membrane bound murine TNF-alpha is: MSTESMIRDVELAEEALPQKMGGFQNSRRCLCLSLFSFLLVAGATTLFCLLNFGVIGPQRDEKFPN GLPLISSMAQTLTLRSSSQNSSDKPVAHVVANHQVEEQLEWLSQRANALLANGMDLKDNQLVV PADGLYLVYSQVLFKGQGCPDYVLLTHTVSRFAISYQEKVNLLSAVKSPCPKDTPEGAELKPWY EPIYLGGVFQLEKGDQLSAEVNLPKYLDFAESGQVYFGVIAL (SEQ ID NO:2). Soluble murine TNF alpha contains amino acids 80-235 ofSEQ ID NO:2.
In some embodiments, the anti-TNF-alpha antibody or antigen-binding fragment thereof binds to human TNF-alpha. In some embodiments, the NF-alpha antibody or antigen-binding fragment thereof is human, humanized, or chimeric.
In some embodiments, the anti-TNF-alpha antibody or antigen-binding fragment thereof binds to murine pha. In some ments, the anti-TNF-alpha antibody or antigen-binding fragment thereof is murine.
In certain embodiments, the anti-TNF-alpha antibody or antigen-binding fragment has one or more of the following effects: neutralizes human pha cytotoxicity in a in vitro L929 assay with an IC50 of 1x10 7 Mor less; blocks the interaction of pha with p55 and p75 cell surface receptors; and/or lyses surface TNF sing cells in vitro in the presence of complement.
In n embodiments, the anti-TNF-alpha antibody or antigen-binding fragment does not bind to TNF -beta.
Anti-TNF-alpha antibodies and antigen-binding fragments thereof include, for example, adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab. Additional anti-TNF-alpha antibodies and antigen-binding fragments are provided, for e, in incorporated by reference in its entirety.
] Adalimumab is bed in U.S. Patent No. 6,258,562, which is herein incorporated by reference in its entirety. Infliximab is described in U.S. Patent No. 5,656,272, which is herein incorporated by reference in its entirety. Certolizumab is discussed in WO 01/94585, which is herein incorporated by reference in its entirety. omab (also known as ) is discussed in Vincent, Int. J Clin. Pract. 54: 190-193 (2000), which is herein orated by reference in its entirety.
Ozoralizumab (also known as ATN-103) is a nanobody. It contains three heavy chain le regions fused by GlySer linkers. Variable regions 1 and 3 are identical, and ozoralizumab does not contain a heavy chain. Ozoralizumab is discussed in in its entirety. Placulumab (also known as CEP-37247) is a domain antibody consisting of a dimer of VL­ pCH l-CH2-CH3 or [V-kappa]2-Fc and is discussed in Gay et al., Mabs 2: 625-638 , which is herein incorporated by reference in its entirety. Golimumab (also known as CNTO 148) is discussed in WO2013/087912, and sequences are provided in GenBank: DI496971. l and GenBank DI .1, each of which is herein incorporated by reference in its entirety.
Anti-TNF-alpha dies and antigen-binding fragments thereof also include antibodies and antigen-binding fragments thereof that competitively inhibit binding of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, or golimumab to TNF-alpha.
Anti-TNF-alpha antibodies and antigen-binding fragments thereof also include antibodies and antigen- g fragments that bind to the same TNF-alpha epitope as adalimumab, infliximab, certolizumab pegol, omab, nerelimomab, ozoralizumab, placulumab, or golimumab.
In n embodiments, the anti-TNF-alpha antibody or antigen-binding fragment thereof competitively inhibits binding of adalimumab to TNF-alpha. In certain embodiments, the NF-alpha antibody or antigen-binding fragment thereof binds to the same TNF-alpha epitope as adalimumab. In certain embodiments, the anti-TNF-alpha antibody or antigen-binding fragment thereof is adalimumab or an antigen-binding fragment thereof. In certain ments, the anti-TNF-alpha antibody or antigenbinding fragment thereof is adalimumab.
In certain embodiments, an anti-TNF-alpha antibody or antigen-binding fragment thereof compnses sequences of adalimumab, imab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, or golimumab, e.g., the complementarity-determining regions (CDRs), the variable heavy domain (VH), and/or the variable light domain (VL). Sequences of exemplary anti-TNF­ alpha antibodies or antigen-binding fragments f are provided in Tables 1-6.
. . T bl 1 Va e ana e eavy c ambl h h CDR ammo ac1 sequences: Antibody VH-CDRl VH-CDR2 VH-CDR3 adalimumab SEQ ID AITWNSGHIDYADSVEG VSYLSTASS(SEQ ID NO:5) NO:3) or (SEQ ID NO:4) VSYLSTASSLDY(SEQ ID GFTFDDYAMH(SEQ NO:94) ID NO:6) infliximab HWMN(SEQ EIRSKSINSATHYAESVKG NYYGSTYDY(SEQ ID ID NO:7) (SEQ ID NO:8) NO:9) certolizumab DYGMN(SEQ ID WINTYIGEPIYADSVKG MDY(SEQ ID NO:10) or (SEQ ID NO:11) NO:12) GYVFTDYGMN(SEQ ID NO:13) afelimomab DYGVN (SEQ ID MIWGDGSTDYDSTLKS EWHHGPVA Y(SEQ ID NO:14) (SEQ ID NO:15) NO:16) momab DYNVD(SEQ ID NINPNNGGTIYNQKFKG SAFYNNYEYFDV(SEQ ID NO:17) (SEQ ID NO:18) NO:19) ozoralizumab Vl:DYWMY(SEQ ID Vl: Vl: R(SEQ ID NO:20) EINTNGLITKYPDSVKG NO:22) V2: SFGMS(SEQ ID (SEQ ID NO:21) V2: GGSLSRSS(SEQ ID NO:23) V2: NO:25) V3: DYWMY (SEQID SISGSGSDTLYADSVKG V3: SPSGFNR(SEQ ID NO:26) (SEQ ID NO:24) NO:28) EINTNGLITKYPDSVKG (SEQ ID NO:27) golimumab GFIFSSYAMH(SEQ FMSYDGSNKKYADSVKG DRGIAAGGNYYYYGMDV ID NO:29) (SEQ ID NO:30) (SEQ ID NO:31) placulumab RASQAIDSYLH(SEQ SASNLET(SEQ ID NO:89) QQVVWRPFT(SEQ ID ID NO:88) NO:90) T bl 2 V . bl l" h h . CDRa e : ana e 1g t c am ammo ac1 ces"d An tibody VL-CDRl VL-CDR2 VL-CDR3 adalimumab RASQGIRNYLA (SEQ ID AASTLQS (SEQ ID PYT (SEQ ID NO:32) NO:33) NO:34) infliximab RASQFVGSSIH (SEQ ID YASESMS (SEQ ID QQSHSWPFT (SEQ ID NO:35) NO:36) NO:37) certolizumab KASQNVGTNVA (SEQ SASFLYS (SEQ ID PLT (SEQ ID ID NO:38) NO:39) NO:40) afelimomab KASQAVSSAVA (SEQ ID WASTRHT (SEQ ID QQHYSTPFT (SEQ ID NO:41) NO:42) NO:43) nerelimomab KSSQSLLYSNNQKNYLA WASTRES (SEQ ID QQYYDYPWT (SEQ ID (SEQ ID NO:44) NO:45) NO:46) ozoralizumab NIA NIA NIA golimumab YSYLA (SEQ ID DASNRAT (SEQ ID PPFT (SEQ ID NO:47) NO:48) NO:49) T bl 3 V . bl ha e : ana e eavy c am ammo ac1 sequencesh.
Antibody VH Am ino Acid Sequence (SEQ ID NO) adalimumab EVQLVESGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWVSA ITWNSGHIDYADSVEGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAKVSY LSTASSLDYWGQGTLVTVSS(SEQ ID NO:50) infliximab SGGGLVQPGGSMKLSCVASGFIFSNHWMNWVRQSPEKGLEWVAE IRSKSINSATHYAESVKGRFTISRDDSKSAVYLQMTDLRTEDTGVYYCSRNY YGSTYDYWGQGTTLTVSS (SEQ ID NO:91) EVKLEESGGGLVQPGGSMKLSCVASGFIFSNHWMNWVRQSPEKGLEWVAE IRSKSINSATHYAESVKGRFTISRDDSKSAVYLQMNSLRTEDTGVYYCSRNY YGSTYDYWGQGTTLTVS(SEQ ID NO:51) certolizumab EVQLVESGGGLVQPGGSLRLSCAASGYVFTDYGMNWVRQAPGKGLEWMG WINTYIGEPIYADSVKGRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCARGY RSYAMDYWGQGTLVTVSS(SEQ ID NO:52) afelimomab QVQLKESGPGLVAPSQSLSITCTVSGFSLTDYGVNWVRQPPGKGLEWLGMI WGDGSTDYDSTLKSRLSISKDNSKSQIFLKNNSLQTDDTARYYCAREWHHG PVAYWGQGTLVTVSA (SEQ ID NO:53) momab QVQLVQSGAEVVKPGSSVKVSCKASGYTFTDYNVDWVKQAPGQGLQWIG NINPNNGGTIYNQKFKGKGTL TVDKSTSTAYMELSSLTSEDTAVYYCARSAF YNNYEYFDVWGQGTTVTVSS(SEQ ID NO:54) ozoralizumab Vl : EVQLVESGGGLVQPGGSLRLSCAASGFTFSDYWMYWVRQAPGKGLEWVSE INTNGLITKYPDSVKGRFTISRDNAKNTLYLQMNSLRPEDTAVYYCARSPSG FNRGQGTLVTVSS (SEQ ID NO:55) EVQLVESGGGLVQPGNSLRLSCAASGFTFSSFGMSWVRQAPGKGLEWVSSI SGSGSDTLYADSVKGRFTISRDNAKTTLYLQMNSLRPEDTAVYYCTIGGSLS RSSQGTLVTVSS (SEQ ID NO:56) EVQLVESGGGLVQPGGSLRLSCAASGFTFSDYWMYWVRQAPGKGLEWVSE INTNGLITKYPDSVKGRFTISRDNAKNTLYLQMNSLRPEDTAVYYCARSPSG FNRGQGTLVTVSS (SEQ ID NO:57) golimumab QVQLVESGGGVVQPGRSLRLSCAASGFIFSSYAMHWVRQAPGNGLEWVAF MSYDGSNKKYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARDR GIAAGGNYYYYGMDVWGQGTTVTVSS(SEQ ID NO:58) T bl 4 V . bl l" ha e : ana e I.Q t c am ammo ac1h . "d ces Antibody VL Amino Acid Sequence (SEQ ID NO) adalimumab DIQMTQSPSSLSASVGDRVTITCRASQGIRNYLAWYQQKPGKAPKLLIYAAS TLQSGVPSRFSGSGSGTDFTLTISSLQPEDVATYYCQRYNRAPYTFGQGTKV EIK (SEQ ID NO:59) infliximab DILLTQSPAILSVSPGERVSFSCRASQFVGSSIHWYQQRTNGSPRLLIKYASES MSGIPSRFSGSGSGTDFTLSINTVESEDIADYYCQQSHSWPFTFGSGTNLEVK (SEQ ID NO:60) certolizumab DIQMTQSPSSLSASVGDRVTITCKASQNVGTNVAWYQQKPGKAPKALIYSA SFLYSGVPYRFSGSGSGTDFTLTISSLQPEDFATYYCQQYNIYPLTFGQGTKV EIK (SEQ ID NO:61 ) omab DIVMTQSHKFMSTTVGDRVSITCKASQAVSSAVAWYQQKPGQSPKLLIYWA STRHTGVPDRFTGSGSVTDFTLTIHNLQAEDLALYYCQQHYSTPFTFGSGTK LEIK (SEQ ID NO:62) nerelimomab DIMMTQSPSTLSASVGDRVTITCKSSQSLLYSNNQKNYLAWYQQKPGQAPK LLISWASTRESGVPSRFIGSGSGTEFTLTISSLQPDDVATYYCQQYYDYPWTF GQGTKVEIK (SEQ ID NO:92) DIMMTQSPSTLSASVGDRVTITCKSSQSLLYSNNQKNYLAWYQQKPGQAPK STRESGVPSRFIGSGSGTEFTLTISSLQPDDVATYYCQQYYDYPWTF GQGTKVEIKR (SEQ ID NO:63) placulumab DIQMTQSPSSLSASVGDRVTITCRASQAIDSYLHWYQQKPGKAPKLLIYSAS NLETGVPSRFSGSGSGTDFTLTISSLLPEDFATYYCQQVVWRPFTFGQGTKV EIK (SEQ ID NO:64) golimumab EIVLTQSPATLSLSPGERATLSCRASQSVYSYLAWYQQKPGQAPRLLIYDAS NRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNWPPFTFGPGTKV DIK (SEQ ID NO:65) T bl 5 F 11 1a e : u - ength heavy c am ammo ac1h. sequences Antibody ength Heavy Chain Amino Acid Sequence (SEQ ID NO) Adalimumab EVQLVESGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWVSA (D2E7) ITWNSGHIDYADSVEGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAKVSY LSTASSLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNV NHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMIS RTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVS VLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSR DELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:66) infliximab EVKLEESGGGLVQPGGSMKLSCVASGFIFSNHWMNWVRQSPEKGLEWVAE IRSKSINSATHYAESVKGRFTISRDDSKSAVYLQMTDLRTEDTGVYYCSRNY YGSTYDYWGQGTTLTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFP EPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVN HKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVL TVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDEL TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKL TVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:67) certolizumab EVQLVESGGGLVQPGGSLRLSCAASGYVFTDYGMNWVRQAPGKGLEWMG WINTYIGEPIYADSVKGRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCARGY RSYAMDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNV NHKPSNTKVDKKVEPKSCDKTHTCAA (SEQ ID NO:68) afelimomab QVQLKESGPGLVAPSQSLSITCTVSGFSLTDYGVNWVRQPPGKGLEWLGMI WGDGSTDYDSTLKSRLSISKDNSKSQIFLKNNSLQTDDTARYYCAREWHHG PVAYWGQGTLVTVSAATTTAPSVYPLVPGCSDTSGSSVTLGCLVKGYFPEP YGALSSGVRTVSSVLQSGFYSLSSLVTVPSSTWPSQTVICNVAHPA SKTELIKRIEPRIPKPSTPPGSSCPPGNILGGPSVFIFPPKPKDALMISLTPKVTC VVVDVSEDDPDVHVSWFVDNKEVHTAWTQPREAQYNSTFRVVSALPIQHQ DWMRGKEFKCKVNNKALPAPIERTISKPKGRAQTPQVYTIPPPREQMSKKK VSLTCLVTNFFSEAISVEWERNGELEQDYKNTPPILDSDGTYFLYSKLTVDT EIFTCSVVHEALHNHHTQKNLSRSPGK (SEQ ID NO:69) ozoralizumab EVQLVESGGGLVQPGGSLRLSCAASGFTFSDYWMYWVRQAPGKGLEWVSE INTNGLITKYPDSVKGRFTISRDNAKNTLYLQMNSLRPEDTAVYYCARSPSG FNRGQGTLVTVSSggggsgggsEVQLVESGGGLVQPGNSLRLSCAASGFTFSSF GMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGRFTISRDNAKTTLYLQM NSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSggggsgggsEVQLVESGGGLVQP GGSLRLSCAASGFTFSDYWMYWVRQAPGKGLEWVSEINTNGLITKYPDSV SRDNAKNTLYLQMNSLRPEDTAVYYCARSPSGFNRGQGTLVTVSS (SEQ ID NO:70) placulumab VEPKSSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVV VDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTV LHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSR DELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDG SFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID N0:93) SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCV VVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLT VLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPS RDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSD GSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:71 ) golimumab QVQLVESGGGVVQPGRSLRLSCAASGFIFSSYAMHWVRQAPGNGLE WVAFMSYDGSNKKYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTA VYYCARDRGIAAGGNYYYYGMDVWGQGTTVTVSSASTKGPSVFPL APSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ SSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKT HTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPE VKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGK EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSL TCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:72) T bl 6 F 11 1a e : u - ength l" ht h .1g1 c am ammo ac1"d sequences Antibody Full-length Light Chain Amino Acid Sequence (SEQ ID NO) umab DIQMTQSPSSLSASVGDRVTITCRASQGIRNYLAWYQQKPGKAPKLLIYAAS (D2E7) TLQSGVPSRFSGSGSGTDFTLTISSLQPEDVATYYCQRYNRAPYTFGQGTKV EIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC (SEQ ID NO:73) infliximab DILLTQSPAILSVSPGERVSFSCRASQFVGSSIHWYQQRTNGSPRLLIKYASES MSGIPSRFSGSGSGTDFTLSINTVESEDIADYYCQQSHSWPFTFGSGTNLEVK RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGN SQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFN RGEC (SEQ ID NO:74) certolizumab DIQMTQSPSSLSASVGDRVTITCKASQNVGTNVAWYQQKPGKAPKALIYSA VPYRFSGSGSGTDFTLTISSLQPEDFATYYCQQYNIYPLTFGQGTKV EIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC (SEQ ID NO:75) afelimomab DIVMTQSHKFMSTTVGDRVSITCKASQAVSSAVAWYQQKPGQSPKLLIYWA STRHTGVPDRFTGSGSVTDFTLTIHNLQAEDLALYYCQQHYSTPFTFGSGTK LEIKRADAAPTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGSERQ NGVLNSWTDQDSKDSTYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKS FNRNEC (SEQ ID NO:76) placulumab DIQMTQSPSSLSASVGDRVTITCRASQAIDSYLHWYQQKPGKAPKLLIYSAS PSRFSGSGSGTDFTLTISSLLPEDFATYYCQQVVWRPFTFGQGTKV EIKR (SEQ ID NO:77) golimumab EIVLTQSPATLSLSPGERATLSCRASQSVYSYLAWYQQKPGQAPRLLIYDAS NRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNWPPFTFGPGTKV DIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC (SEQ ID NO:78) Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 80% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively. or Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 85% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 85% sequence identity to SEQ ID NOs: 91 and 60, or 54 and 92, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 90% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively. Also provided are antibodies or n-binding fragments that comprise a VH and a VL having at least 90% sequence identity to SEQ ID NOs: 91 and 60, or 54 and 92, respectively. Also ed are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 95% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 95% ce identity to SEQ ID NOs: 91 and 60, or 54 and 92, tively. Also ed are antibodies or n-binding fragments that comprise a VH and a VL having at least 96% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, tively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 96% sequence identity to SEQ ID NOs: 91 and 60, or 54 and 92, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 97% ce identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 97% sequence identity to SEQ ID NOs: 91 and 60, or 54 and 92, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 98% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively. Also provided are antibodies or n-binding fragments that comprise a VH and a VL having at least 98% ce identity to SEQ ID NOs: 91 and 60, or 54 and 92, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 99% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 99% sequence identity to SEQ ID NOs: 91 and 60, or 54 and 92, respectively.
Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 80% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively, and n the CDRs of SEQ ID NOs: 3 or 6, 4, 5, and 32-34; 7-9 and 35- 37; 10 or 13, 11, 12, and 38-40; 14-16, and 41-43; 17-19 and 44-46; or 29-31 and 47-49, respectively.
Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 80% ce identity to SEQ ID NOs: 50 and 59, 91 and 60, or 54 and 92 respectively, and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 94, and 32-34; 7-9 and 35-37; or 17-19 and 44-46, tively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 85% ce identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, tively and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 5, and 32-34; 7-9 and 35-37; 10 or 13, 11, 12, and 38-40; 14-16, and 41-43; 17-19 and 44-46; or 29-31 and 47-49, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 85% sequence ty to SEQ ID NOs: 50 and 59, 91 and 60, or 54 and 92 respectively, and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 94, and 32-34; 7-9 and 35-37; or 17-19 and 44-46, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 90% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 5, and 32-34; 7-9 and 35-37; 10 or 13, 11, 12, and 38-40; 14-16, and 41-43; 17-19 and 44-46; or 29-31 and 47-49, respectively. Also provided are dies or antigen-binding fragments that comprise a VH and a VL having at least 90% sequence identity to SEQ ID NOs: 50 and 59, 91 and 60, or 54 and 92 respectively, and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 94, and 32-34; 7-9 and 35-37; or 17-19 and 44-46, respectively. Also provided are antibodies or antigen-binding nts that comprise a VH and a VL having at least 95% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 5, and 32-34; 7-9 and 35-37; 10 or 13, 11, 12, and 38-40; 14-16, and 41-43; 17-19 and 44-46; or 29-31 and 47-49, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 95% sequence identity to SEQ ID NOs: 50 and 59, 91 and 60, or 54 and 92 respectively, and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 94, and 32-34; 7-9 and 35-37; or 17-19 and 44-46, respectively. Also provided are dies or antigen-binding fragments that comprise a VH and a VL having at least 96% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively and n the CDRs of SEQ ID NOs: 3 or 6, 4, 5, and 32-34; 7-9 and 35-37; 10 or 13, 11, 12, and 38-40; 14-16, and 41-43; 17-19 and 44-46; or 29-31 and 47-49, respectively. Also ed are antibodies or antigen-binding fragments that se a VH and a VL having at least 96% sequence identity to SEQ ID NOs: 50 and 59, 91 and 60, or 54 and 92 respectively, and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 94, and 32-34; 7-9 and 35-37; or 17-19 and 44-46, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 97% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively and contain the CDRs of SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively. Also provided are dies or antigen-binding fragments that comprise a VH and a VL having at least 97% ce identity to SEQ ID NOs: 50 and 59, 91 and 60, or 54 and 92 respectively, and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 94, and 32-34; 7-9 and 35-37; or 17-19 and 44-46, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 98% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively and n the CDRs of SEQ ID NOs: 3 or 6, 4, 5, and 32-34; 7-9 and -37; 10 or 13, 11, 12, and 38-40; 14-16, and 41-43; 17-19 and 44-46; or 29-31 and 47-49, respectively.
Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 98% ce identity to SEQ ID NOs: 50 and 59, 91 and 60, or 54 and 92 respectively, and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 94, and 32-34; 7-9 and 35-37; or 17-19 and 44-46, respectively. Also provided are dies or antigen-binding fragments that comprise a VH and a VL having at least 99% sequence identity to SEQ ID NOs: 50 and 59, 51 and 60, 52 and 61, 53 and 62, 54 and 63, or 58 and 65, respectively and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 5, and 32-34; 7-9 and 35-37; 10 or 13, 11, 12, and 38-40; 14-16, and 41-43; 17-19 and 44-46; or 29-31 and 47-49, respectively. Also provided are antibodies or antigen-binding fragments that comprise a VH and a VL having at least 99% sequence identity to SEQ ID NOs: 50 and 59, 91 and 60, or 54 and 92 respectively, and contain the CDRs of SEQ ID NOs: 3 or 6, 4, 94, and 32-34; 7-9 and 35-37; or 17-19 and 44-46, respectively.
In certain embodiments, the anti-TNF alpha antibody or antigen-binding fragment thereof comprises the CDRs of SEQ ID NOs: 3-5 and 32-34 or of SEQ ID NOs: 6, 4, 5, and 32-34. In certain embodiments, the anti-TNF alpha antibody or antigen-binding nt thereof ses the CDRs of SEQ ID NOs: 3, 4, 94 and 32-34 or of SEQ ID NOs: 6, 4, 94, and 32-34. In certain embodiments, the anti-TNF alpha antibody or antigen-binding fragment thereof comprises the VH of SEQ ID NO:50 and/or the VL of SEQ ID N0:59. In certain embodiments, the anti-TNF alpha antibody comprises the heavy chain of SEQ IDNO: 66 and/or the light chain of SEQ ID N0:75.
F alpha antibody ses the heavy chain of SEQ ID N0:74 and/or the light chain of SEQ ID N0:82.
In certain aspects, provided herein are antibodies or n-binding fragments thereof that specifically bind to TNF-alpha and comprise the Chothia VL CDRs of a VL of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, or golimumab. In certain aspects, provided herein are antibodies or antigen-binding fragments thereof that specifically bind to TNF­ alpha and comprise the Chothia VH CDRs of a VH of adalimumab, infliximab, certolizumab pegol, afelimomab, momab, izumab, placulumab, or golimumab. In certain aspects, provided herein are antibodies or antigen-binding fragments thereof that specifically bind to TNF-alpha and comprise the Chothia VL CDRs of a VL of umab, infliximab, izumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, or golimumab and comprise the Chothia VH CDRs of a VH of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, or mab. In n embodiments, antibodies or antigen-binding fragments that specifically bind to TNF-alpha comprise one or more CDRs, in which the Chothia and Kabat CDRs have the same amino acid sequence.
In n embodiments, provided herein are antibodies and antigen-binding fragments thereof that specificallybind to TNF-alpha and comprise combinations of Kabat CDRs and Chothia CDRs.
In a particular embodiment, provided herein are antibodies or antigen-binding fragments thereof that specifically bind to TNF-alpha and se CDRs of adalimumab, infliximab, izumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, or golimumab as determined by the IMGT numbering system, for example, as described in Lefranc M-P (1999) supra and c M-P et al., (1999) supra).
In a particular embodiment, ed herein are dies that specifically bind to TNF- alpha and comprise CDRs of umab, infliximab, certolizumab pegol, afelimomab, momab, ozoralizumab, placulumab, or golimumab as determined by the method in MacCallum RM et al.
In a particular ment, provided herein are antibodies or antigen-binding fragments thereof that specifically bind to TNF-alpha and comprise CDRs of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, or golimumab as determined by the AbM numbering scheme.
In a particular embodiment, provided herein are antibodies or antigen-binding nts thereof that specifically bind to CD163.
Monoclonal antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein (1975) Nature 256:495. Using the hybridoma method, a mouse, hamster, or other appropriate host animal, is immunized to elicit the production by lymphocytes of antibodies that will specifically bind to an immunizing antigen. Lymphocytes can also be immunized in vitro. Following immunization, the lymphocytes are isolated and fused with a suitable myeloma cell line using, for example, polyethylene glycol, to form hybridoma cells that can then be selected away from unfused lymphocytes and a cells. Hybridomas that e monoclonal antibodies directed specifically t a chosen antigen as determined by immunoprecipitation, immunoblotting, or by an in vitro binding assay (e.g., radioimmunoassay (RIA); enzyme-linked immunosorbent assay )) can then be propagated either in vitro culture using standard methods (Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, 1986) or in vivo as ascites tumors in an animal. The onal antibodies can then be purified from the culture medium or ascites fluid as described for polyclonal antibodies.
] Alternatively monoclonal antibodies can also be made using recombinant DNA s as described in U.S. Patent 4,816,567. The polynucleotides encoding a monoclonal antibody are isolated from mature B-cells or hybridoma cells, such as by RT-PCR using oligonucleotide primers that specifically y the genes encoding the heavy and light chains of the antibody, and their sequence is determined using conventional procedures. The isolated polynucleotides encoding the heavy and light chains are then cloned into suitable expression vectors, which when transfected into host cells such as E. coli cells, simian COS cells, e hamster ovary (CHO) cells, or a cells that do not ise produce immunoglobulin protein, monoclonal antibodies are generated by the host cells. Also, recombinant monoclonal antibodies or fragments thereof of the desired species can be isolated from phage display libraries expressing CDRs of the desired species as described ferty et al., 1990, Nature, 348:552-554; Clackson et al., 1991, , 352:624-628; and Marks et al., 1991, J. Mol. Biol., 222:581- 597).
] The polynucleotide(s) encoding a monoclonal dy can further be modified in a number of different manners using recombinant DNA technology to generate ative antibodies. In some embodiments, the constant domains of the light and heavy chains of, for example, a mouse onal antibody can be substituted 1) for those regions of, for example, a human antibody to generate a chimeric antibody or 2) for a non-immunoglobulin polypeptide to generate a fusion antibody. In some embodiments, the constant regions are truncated or removed to generate the desired antibody fragment of a monoclonal antibody. irected or high-density mutagenesis of the variable region can be used to optimize specificity, affinity, etc. of a monoclonal antibody.
In some embodiments, the monoclonal antibody against the TNF-alpha is a humanized antibody. In certain embodiments, such antibodies are used therapeutically to reduce antigenicity and HAMA (human anti-mouse antibody) ses when administered to a human subject.
Methods for engineering, zing or resurfacing non-human or human antibodies can also be used and are well known in the art. A humanized, resurfaced or similarly engineered antibody can have one or more amino acid residues from a source that is non-human, e.g., but not limited to, mouse, rat, , man primate or other mammal. These non-human amino acid residues are replaced by residues that are often referred to as "import" residues, which are typically taken from an "import" variable, constant or other domain of a known human sequence.
Such imported sequences can be used to reduce immunogenicity or reduce, enhance or modify binding, affinity, on-rate, off-rate, avidity, specificity, half-life, or any other suitable characteristic, as known in the art. In general, the CDR residues are directly and most substantially ed in influencing TNF-alpha binding. Accordingly, part or all of the non-human or human CDR sequences are maintained while the non-human sequences of the variable and nt regions can be replaced with human or other amino acids.
Antibodies can also optionally be humanized, resurfaced, engineered or human antibodies engineered with retention of high affinity for the antigen e.g., pha, and other favorable biological properties. To achieve this goal, humanized (or human) or engineered antibodies and resurfaced antibodies can be optionally prepared by a process of analysis of the parental sequences and various conceptual humanized and ered ts using three-dimensional models of the parental, engineered, and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these ys permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen, such as TNF-alpha. In this way, ork (FR) es can be selected and combined from the consensus and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved.
Humanization, resurfacing or engineering of antibodies of the present disclosure can be performed using any known , such as but not limited to those described in, Winter (Jones et al., Nature 2 (1986); Riechmann et al., Nature 332:323 (1988); Verhoeyen et al., Science 34 (1988)), Sims et al., J. Immunol. 151: 2296 (1993); Chothia and Lesk, J. Mol. Biol. 196:901 (1987), Carter et al., Proc. Natl. Acad. Sci. U.S.A. 89:4285 (1992); Presta et al., J. Immunol. 151:2623 (1993), U.S. Pat. Nos. 5,639,641, 323; 5,976,862; 5,824,514; 5,817,483; 5,814,476; 5,763,192; 5,723,323; ,766,886; 5,714,352; 6,204,023; 6,180,370; 762; 5,530,101; 5,585,089; 5,225,539; 4,816,567; PCT/: US98/16280; US96/18978; US91/09630; US91/05939; US94/01234; GB89/01334; GB91/0l 134; GB92/0l 755; WO90/14443; 4424; WO90/14430; EP 229246; 7,557,189; 7,538,195; and 7,342,110, each of which is entirely incorporated herein by reference, including the references cited therein.
In n ative embodiments, the antibody (e.g., an anti-TNFalpha antibody) is a human antibody. Human antibodies can be directly ed using various techniques known in the art.
Immortalized human B lymphocytes immunized in vitro or isolated from an immunized individual that produce an antibody directed against a target antigen can be generated (See, e.g., Cole et al., onal dies and Cancer Therapy, Alan R. Liss, p. 77 (1985); Boemer et al., 1991, J. Immunol., 147 (1):86- 95; and U.S. Patent 5,750,373). Also, the human antibody can be selected from a phage library, where that phage library expresses human dies, as described, for example, in Vaughan et al., 1996, Nat.
Biotech., 14:309-314, Sheets et al., 1998, Proc. Nat'l. Acad. Sci., 95:6157-6162, Hoogenboom and Winter, 1991, J. Mol. Biol., 227:381, and Marks et al., 1991, J. Mol. Biol., 222:581). Techniques for the tion and use of antibody phage libraries are also described in U.S. Patent Nos. 5,969,108, 197, 5,885,793, 6,521,404; 6,544,731; 6,555,313; 6,582,915; 6,593,081; 6,300,064; 6,653,068; 6,706,484; and 7,264,963; and Rothe et al., 2007, J. Mol. Bio., doi:10.1016/j.jmb.2007.12.018 (each of which is incorporated by reference in its entirety). Affinity maturation strategies and chain ing strategies (Marks et al., 1992, Bio/Technology 10:779-783, incorporated by reference in its entirety) are known in the art and can be employed to generate high affinity human antibodies.
Humanized antibodies can also be made in transgenic mice ning human immunoglobulin loci that are capable upon immunization of producing the full repertoire of human antibodies in the absence of endogenous immunoglobulin production. This approach is described in U.S.
Patents 5,545,807; 5,545,806; 5,569,825; 5,625,126; 425; and 016.
In certain ments are provided an antibody fragment to, for example, increase tumor penetration. Various techniques are known for the production of antibody fragments. Traditionally, these fragments are derived via proteolytic ion of intact dies (for example Morimoto et al., 1993, Journal of Biochemical and sical Methods 24:107-117; Brennan et al., 1985, Science, 229:81). In certain embodiments, antibody fragments are produced recombinantly. Fab, Fv, and scFv antibody fragments can all be expressed in and secreted from E. coli or other host cells, thus allowing the production of large s of these fragments. Such antibody fragments can also be isolated from antibody phage libraries. The antibody fragment can also be linear antibodies as described in U.S. Patent ,641,870. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner.
For the purposes of the present disclosure, it should be appreciated that modified antibodies can comprise any type of variable region that provides for the ation of the antibody with the antigen (e.g., TNF alpha). In this regard, the variable region can comprise or be derived from any type of mammal that can be induced to mount a l se and generate immunoglobulins against the desired tumor associated antigen. As such, the variable region of the modified antibodies can be, for example, of human, murine, non-human e (e.g., cynomolgus monkeys, macaques, etc.) or lupine origin. In some embodiments both the variable and constant regions of the modified immunoglobulins are human. In other embodiments the variable regions of compatible antibodies (usually derived from a non- human ) can be engineered or specifically tailored to improve the binding properties or reduce the immunogenicity of the le. In this respect, variable regions useful in the present disclosure can be humanized or otherwise altered through the inclusion of imported amino acid sequences.
In certain embodiments, the variable domains in both the heavy and light chains are d by at least partial replacement of one or more CDRs and, if necessary, by partial framework region replacement and sequence changing. Although the CDRs can be derived from an antibody of the same class or even subclass as the antibody from which the framework s are derived, it is envisaged that the CDRs will be d from an antibody of different class and in certain embodiments from an antibody from a different species. It may not be necessary to replace all of the CDRs with the complete CDRs from the donor variable region to transfer the antigen-binding capacity of one variable domain to another. , it may only be necessary to transfer those residues that are necessary to maintain the activity of the antigen-binding site. Given the explanations set forth in U.S. Pat. Nos. 5,585,089, ,693,761 and 5,693,762, it will be well within the ence of those skilled in the art, either by carrying out routine experimentation or by trial and error testing to obtain a functional antibody with reduced immunogenicity.
] Anti-TNF alpha proteins include soluble TNF receptor proteins. The NF alpha protein can be a soluble p75 TNF receptor. The anti-TNF alpha protein can be a e p55 TNF or.
The soluble TNF receptor can bind to both TNF alpha and TNF beta. The soluble TNF receptor can bind to TNF alpha, but not to TNF beta.
The soluble TNF receptor can inhibit binding of TNF alpha (and optionally TNF beta) to cell surface TNF ors.
The soluble TNF receptor can be etanercept.
An anti-TNF alpha protein, e.g., a soluble TNF receptor, can be fused to a heavy chain constant domain or fragment thereof or an Fe region or fragment thereof. The heavy chain constant domain fragment or Fe fragment can be a portion of the constant domain or Fe that is capable of binding to Fe receptor. The heavy chain constant domain fragment or Fe fragment can be a portion of the constant domain or Fe that is capable of inducing cell lysis in vitro in the presence of complement. The heavy chain constant domain fragment or Fe fragment can be a portion of the constant domain or Fe that is capable of inducing ADCC.
The heavy chain constant domain or fragment f or Fe region or fragment thereof can be a human heavy chain constant domain or fragment thereof or human Fe region or fragment thereof. The heavy chain constant domain or fragment thereof or Fe region or fragment thereof can be an IgG 1 heavy chain constant domain or nt thereof or an IgG 1 Fe region or fragment f. The heavy chain constant domain or fragment thereof or Fe region or fragment thereof can be a human IgG 1 heavy chain constant domain or fragment thereof or human IgG1 F c region or nt thereof.
Those skilled in the art will appreciate that the antibodies and antigen-binding fragments thereof of this disclosure and the anti-TNF proteins of this disclosure e dies, antigen-binding fragments thereof, and anti-TNF proteins (e.g., full-length antibodies, antigen-binding fragments of dies, or soluble TNF receptor proteins) comprising one or more of constant region domains, including domains that have been altered so as to provide desired biochemical characteristics such as reduced serum ife when compared with an antibody, n-binding fragment thereof, or anti-TNF protein of approximately the same immunogenicity comprising a native or unaltered constant . In some embodiments, the constant region of the antibody, n-binding fragment thereof, or anti-TNF protein (e.g., full-length antibodies, antigen-binding fragments of antibodies, or soluble TNF receptor proteins) will comprise a human constant region. Modifications to the constant region compatible with this sure comprise additions, deletions, or substitutions of one or more amino acids in one or more domains. That is, the antibody, antigen-binding fragment thereof, or anti-TNF proteins (e.g., full-length antibodies, antigen-binding fragments of antibodies, or soluble TNF receptor proteins) disclosed herein can comprise alterations or modifications to one or more of the three heavy chain constant domains (CHI, CH2 or CH3) and/or to the light chain constant domain (CL). In some embodiments, modified constant s wherein one or more s are partially or entirely deleted are contemplated. In some embodiments, the antibodies, antigen-binding fragments thereof, or anti-TNF proteins (e.g., full-length antibodies, n-binding fragments of antibodies, or soluble TNF receptor proteins) will comprise domain deleted constructs or ts wherein the entire CH2 domain has been removed (ACH2 constructs). In some embodiments, the omitted constant region domain will be replaced by a short amino acid spacer (e.g., 10 es) that provides some of the molecular flexibility typically imparted by the absent nt region.
It will be noted that m certain embodiments, the dies, antigen-binding fragments thereof, or anti-TNF proteins (e.g., full-length antibodies, antigen-binding fragments of antibodies, or soluble TNF receptor proteins) can be engineered to fuse the CH3 domain directly to the hinge region of the respective antibodies, antigen-binding nts thereof, or anti-TNF ns (e.g., full-length antibodies, antigen-binding fragments of antibodies, or e TNF receptor proteins). In other constructs it can be ble to provide a peptide spacer between the hinge region and the modified CH2 and/or CH3 domains. For example, compatible constructs could be expressed wherein the CH2 domain has been deleted and the remaining CH3 domain (modified or unmodified) is joined to the hinge region with a 5-20 amino acid spacer. Such a spacer can be added, for instance, to ensure that the regulatory elements of the constant domain remain free and accessible or that the hinge region remains le.
However, it should be noted that amino acid spacers can, in some cases, prove to be genic and elicit an unwanted immune response against the construct. Accordingly, in certain embodiments, any spacer added to the construct will be relatively non-immunogenic, or even omitted altogether, so as to maintain the d biochemical qualities of the antibodies, antigen-binding fragments f, or anti­ TNF proteins (e.g., full-length antibodies, antigen-binding fragments of dies, or soluble TNF receptor proteins).
It will be appreciated that the antibodies, antigen-binding nts therof, and anti-TNF proteins (e.g., full-length dies, antigen-binding nts of antibodies, or soluble TNF receptor proteins) of the present disclosure can be provided by the partial deletion or substitution of a few or even a single amino acid. For example, the mutation of a single amino acid in selected areas of the CH2 domain can be enough to substantially reduce Fe g and thereby increase tumor localization. Similarly, it may be desirable to simply delete that part of one or more constant region domains that control the or function (e.g., complement ClQ binding) to be modulated. Such partial deletions of the constant regions can improve selected characteristics of the antibody (serum half-life) while leaving other desirable functions associated with the subject nt region domain intact. Moreover, as alluded to above, the constant regions of the sed dies, antigen-binding nts therof, and anti-TNF proteins (e.g., full-length antibodies, antigen-binding fragments of antibodies, or soluble TNF receptor proteins) can be modified through the mutation or substitution of one or more amino acids that enhances the profile of the resulting uct. In this respect it can be possible to disrupt the activity provided by a conserved binding site (e.g., Fe binding) while substantially maintaining the configuration and immunogenic e of the antibodies, antigen-binding fragments therof, and anti-TNF proteins (e.g., full-length antibodies, antigen-binding fragments of antibodies, or soluble TNF receptor proteins). Certain embodiments can se the addition of one or more amino acids to the constant region to enhance desirable characteristics such as decreasing or increasing effector function or provide for more glucocorticoid receptor agonist attachment. In such embodiments it can be ble to insert or replicate specific sequences derived from selected constant region domains.
It will be appreciated that the antibodies, n-binding fragments therof, and anti-TNF proteins (e.g., ength antibodies, antigen-binding fragments of antibodies, or soluble TNF receptor ns) of the present disclosure can be modified to reduce immunogenicity, i.e., to reduce the anti-drug immune response (ADA). Methods ofdoing so are disclosed, for example, in herein incorporated by reference in its ty.
The present disclosure further embraces variants and equivalents which are substantially homologous to antibodies, antigen-binding fragments therof, and anti-TNF proteins (e.g., full-length antibodies, n-binding fragments of antibodies, or soluble TNF receptor proteins) set forth herein.
These can contain, for example, conservative tution mutations, i.e., the substitution of one or more amino acids by similar amino acids. For example, conservative substitution refers to the substitution ofan amino acid with another within the same general class such as, for example, one acidic amino acid with another acidic amino acid, one basic amino acid with another basic amino acid or one neutral amino acid by another neutral amino acid. What is intended by a conservative amino acid substitution is well known in the art.
The polypeptides of the present disclosure can be recombinant ptides, natural polypeptides, or synthetic polypeptides of an antibody, antigen-binding fragment f, or anti-TNF protein. It will be recognized in the art that some amino acid sequences of the disclosure can be varied without significant effect of the structure or function of the protein. Thus, the disclosure further includes variations of the polypeptides which show substantial ty or which include regions of an antibody, antigen-binding fragment thereof, or anti-TNF alpha protein. Such s include deletions, insertions, inversions, repeats, and type substitutions.
The polypeptides and analogs can be further modified to contain additional chemical moieties not normally part of the protein. Those derivatized moieties can improve the lity, the biological half life or tion of the protein. The moieties can also reduce or eliminate any desirable side effects of the proteins and the like. An overview for those moieties can be found in REMINGTON'S PHARMACEUTICAL SCIENCES, 20th ed., Mack Publishing Co., , PA (2000).
The isolated polypeptides described herein can be produced by any suitable method known in the art. Such methods range from direct protein tic methods to ucting a DNA ce ng isolated polypeptide sequences and expressing those sequences in a suitable transformed host.
In some embodiments, a DNA sequence is constructed using recombinant technology by isolating or synthesizing a DNA sequence encoding a wild-type protein of interest. Optionally, the sequence can be mutagenized by site-specific mutagenesis to provide functional analogs thereof. See, e.g., Zoeller et al., Proc. Nat'l. Acad. Sci. USA 2-5066 (1984) and U.S. Pat. No. 4,588,585.
In some embodiments a DNA sequence encoding a ptide of interest would be constructed by chemical synthesis using an oligonucleotide synthesizer. Such ucleotides can be designed based on the amino acid sequence of the desired ptide and selecting those codons that are favored in the host cell in which the recombinant polypeptide of interest will be produced. Standard methods can be applied to synthesize an isolated polynucleotide sequence encoding an isolated ptide of interest. For example, a complete amino acid sequence can be used to construct a backtranslated gene. Further, a DNA oligomer containing a nucleotide sequence coding for the ular isolated polypeptide can be synthesized. For example, l small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated. The individual oligonucleotides typically contain 5' or 3' ngs for complementary assembly.
Once led (by synthesis, site-directed mutagenesis or another method), the polynucleotide sequences encoding a particular isolated polypeptide of interest will be inserted into an expression vector and operatively linked to an expression control sequence appropriate for expression of the protein in a desired host. Proper assembly can be confirmed by nucleotide sequencing, restriction mapping, and expression of a biologically active polypeptide in a suitable host. As is well known in the art, in order to obtain high expression levels of a transfected gene in a host, the gene must be operatively linked to transcriptional and translational expression control sequences that are functional in the chosen sion host.
In certain embodiments, recombinant expression vectors are used to amplify and express DNA encoding antibodies, antigen-binding fragments thereof, or anti-TNF proteins (e.g., full-length antibodies, antigen-binding nts of antibodies, or soluble TNF receptor proteins). Recombinant sion vectors are replicable DNA constructs which have tic or cDNA-derived DNA fragments encoding a polypeptide chain of an antibody, antigen-binding fragment thereof, or anti-TNF protein (e.g., full-length antibodies, antigen-binding fragments of antibodies, or soluble TNF or proteins), operatively linked to suitable transcriptional or ational regulatory elements derived from mammalian, ial, viral or insect genes. A transcriptional unit lly comprises an assembly of (1)a genetic element or elements having a regulatory role in gene expression, for example, transcriptional promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription and translation initiation and termination sequences. Such regulatory elements can include an operator ce to control transcription. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants can additionally be incorporated. DNA regions are operatively linked when they are functionally related to each other. For example, DNA for a signal peptide (secretory leader) is operatively linked to DNA for a polypeptide if it is expressed as a precursor which participates in the ion of the polypeptide; a promoter is operatively linked to a coding sequence if it controls the transcription of the sequence; or a ribosome binding site is operatively linked to a coding sequence if it is positioned so as to permit translation. Structural elements intended for use in yeast expression systems e a leader sequence enabling extracellular secretion of ated protein by a host cell.
Alternatively, where recombinant n is sed without a leader or transport sequence, it can include an N-terminal methionine residue. This residue can optionally be subsequently cleaved from the expressed inant protein to provide a final product.
The choice of expression control sequence and expression vector will depend upon the choice of host. A wide variety of expression ector combinations can be employed. Useful expression vectors for eukaryotic hosts, include, for example, s comprising expression control ces from SV 40, bovine papilloma virus, irus and cytomegalovirus. Useful expression vectors for bacterial hosts include known bacterial plasmids, such as plasmids from Escherichia coli, including pCR 1, pBR322, pMB9 and their tives, wider host range plasmids, such as M13 and filamentous stranded DNA phages.
Suitable host cells for expression of antibodies, antigen-binding fragments thereof, and anti­ TNF proteins (e.g., full-length antibodies, antigen-binding fragments of antibodies, or soluble TNF receptor proteins) include prokaryotes, yeast, insect or higher eukaryotic cells under the control of appropriate promoters. Prokaryotes include gram ve or gram positive organisms, for example E. coli or bacilli. Higher eukaryotic cells include established cell lines of mammalian origin. Cell-free translation systems could also be employed. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. (Cloning Vectors: A Laboratory Manual, Elsevier, N.Y., 1985), the relevant disclosure of which is hereby incorporated by reference. Additional information regarding methods of protein production, including dy production, can be found, e.g., in U.S. Patent Publication No. 2008/0187954, U.S. Patent Nos. 6,413,746 and 6,660,501, and International Patent ation No. WO 04009823, each of which is hereby incorporated by reference herein in its entirety.
] Various mammalian or insect cell culture s are also advantageously employed to express recombinant n. sion of recombinant proteins in ian cells can be performed because such proteins are generally correctly folded, appropriately modified and completely functional.
Examples of suitable ian host cell lines include HEK-293 and HEK-293T, the COS-7 lines of monkey kidney cells, bed by Gluzman (Cell 23:175, 1981), and other cell lines ing, for example, L cells, Cl27, 3T3, Chinese hamster ovary (CHO), HeLa and BHK cell lines. Mammalian expression vectors can comprise nontranscribed elements such as an origin of replication, a le promoter and enhancer linked to the gene to be expressed, and other 5' or 3' flanking nontranscribed sequences, and 5' or 3' nontranslated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination ces.
Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Bio/Technology 6:47 (1988).
] The proteins produced by a transformed host can be purified according to any suitable method. Such standard methods include chromatography (e.g., ion exchange, affinity and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification. ty tags such as hexahistidine, maltose binding domain, influenza coat sequence and hione-S-transferase can be attached to the protein to allow easy purification by passage over an appropriate affinity column. Isolated proteins can also be ally characterized using such ques as proteolysis, nuclear magnetic resonance and x-ray crystallography.
For example, atants from systems which secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. Following the concentration step, the concentrate can be applied to a suitable purification matrix. Alternatively, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification.
Alternatively, a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising ropyl or carboxymethyl groups. Finally, one or more reversed-phase high performance liquid tography (RP-HPLC) steps employing hydrophobic RP-HPLC media, e.g., silica gel having t methyl or other aliphatic groups, can be employed to further purify anti­ TNF proteins (e.g., full-length antibodies, antigen-binding nts of antibodies, or e TNF receptor proteins). Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a homogeneous recombinant protein.
Recombinant protein produced in bacterial culture can be isolated, for example, by l extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion ge or size exclusion chromatography steps. High mance liquid chromatography (HPLC) can be employed for final purification steps. Microbial cells employed in expression of a recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, ical disruption, or use of cell lysing agents.
Methods known in the art for purifying dies, antigen-binding fragments thereof, and anti-TNF alpha proteins also include, for example, those described in U.S. Patent Publication Nos. 2008/0312425, 2008/0177048, and 2009/0187005, each of which is hereby incorporated by reference herein in its entirety.
III. Immunoconjugates containing glucocorticoid receptor agonists ] Immunoconjugates containing orticoid receptor agonists are provided herein. In some embodiments, an conjugate provided herein binds to Fe gamma receptor. In some embodiments, an conjugate provided herein is active in the GRE transmembrane TNF-alpha reporter assay (as used herein the " GRE transmembrane TNF-alpha reporter assay" refers to the assay used in Example 79 below). In some embodiments, an immunoconjugate provided herein is active in the L929 assay (as used herein, the "L929 assay" refers to the assay used in Example 82 below). In some embodiments, an immunoconjugate provided herein shows reduced immunogenicity (reduced anti-drug immune response (ADA)) as ed to the protein in the immunoconjugate (e.g., the antibody, antigen-binding fragment thereof, or soluble receptor) alone.
In one ment, disclosed herein is a compound having Formula I-a: (SM-L-Q)n-A1 I-a or a pharmaceutically acceptable salt thereof, wherein: A1 is an anti-tumor is factor (TNF) alpha protein; L is a linker; Q is a heterobifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a radical of a glucocorticosteroid.
In one embodiment, disclosed herein is a nd having Formula I-a: (SM-L-Q)n-A1 I-a or a pharma ceutically acceptable salt thereof, wherein: A1 is an anti-tumor necrosis factor (TNF) alpha antibody, an anti-TNF alpha monoclonal antibody, or adalimumab; L is a linker; Q is a bifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a radical of a glucocorticosteroid.
In another ment, disclosed herein 1s a compound having Formula I-a, or a c eutically able salt thereof, wherein SM is a monovalent l of a glucocorticosteroid.
In another ment, disclosed herein is a compound having Formula I-a, or a pharmac eutically acceptable salt thereof, wherein SM is a monovalent l of a glucocorticosteroid selected from the group consisting of: SM S-� , SM O-� , SM N-� and SM N-� wherein the sulfur, oxygen, or nitrogen atom is attached directly or indirectly to the C- or D-ring of the glucocorticosteroid, and R is C1 4 alkyl. In another ment, the sulfur, oxygen, or nitrogen atom is attached directly or indirectly to the D-ring ofthe glucocorticosteroid.
In another embodiment, disclosed herein is a compound having a I-a, or a pharmac lly acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula II-a: II-a, wherein: R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is ed from the group consisting of -CH2OH, -CH2SH, -CH2Cl, -SCH2Cl, -SCH2F, -SCH2CF3, -OH (or hydroxy), -OCH2CN, l, -OCH2F, -OCH3, -OCH2CH3, -SCH2CN, /�o�OH oy•,, o H /�o yo yR 3b /�o YR 3c C02H R3a O 0 R3a is selected from the group consisting ofhydrogen and Ci-4 alkyl; R3b is ed from the group consisting of C1_4 alkyl and Ci-4 alkoxy; R3c is selected from the group consisting ofhydrogen, C1_4 alkyl, , and C1_4 alkoxy; R3ct and R3e are independently selected from hydrogen and Ci-4 alkyl; R9a is selected from the group consisting of optionally substituted alkyl, optionally substituted cycloalkyl, ally substituted aryl, and optionally substituted heteroaryl; R9b is selected from the group consisting ofhydrogen and alkyl; or R9a is: j' "j( \�R6e R6c R11 R6d ; and R9b is hydrogen or methyl; X is selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=O)-, -S(=O)r, -NR5-, -CH2S-, , -N(H)C(R8a)(R8b)-, -CR4c=CR4ct-, and -C=C-; or Xis absent; tis I or 2; Z is selected from the group ting , =C(OH)-, and =N-; each R4a and R4b are independently selected from the group consisting of hydrogen and Ci-4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a3- to 6-membered cycloalkyl; R4c and R4ct are ndently selected from the group consisting ofhydrogen and Ci-4 alkyl; R5 is selected from the group consisting ofhydrogen and C1_4 alkyl; R6\ R6\ R6C, R6ct, and R6e are each independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and ; R8a and R8b are independently ed from the group consisting ofhydrogen and Ci-4 alkyl; R11 is selected from the group consisting of hydrogen, halo, C1_4 alkyl, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
In another embodiment, disclosed herein is a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having Formula II-a, wherein R9a is: In another embodiment, disclosed herein is a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having Formula II-a': II-a', wherein R1, R2, R3, R9\ R9b and=-= are as defined in connection with a II-a.
In another embodiment, sed herein is a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having Formula II-b: .,,0--1 R9a ''' /'R9b wherein R1, R2, R3, R9\ R9\ and=-= are as defined in tion with Formula II-a.
In another embodiment, disclosed herein is a nd having a I-a, wherein SM is a monovalent radical of a glucocorticosteroid having Formula II-b': .,,0--1 R9a ''' /'R9b II-b' wherein R1, R2, R3, R9\ R9\ and=-= are as defined in tion with Formula II-a.
In another embodiment, disclosed herein is a nd having Formula I-a, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula II-c: ,.,.\ 11-c, wherein R1, R2, R9\ R9\ and=-= are as defined in connection with Formula II-a; and Wis selected from the group consisting of and -S-. In r embodiment, Wis . In r embodiment, Wis -S-.
In another embodiment, sed herein is a compound having Formula I-a, wherein SM is a monovalent radical ofa glucocorticosteroid having Formula 11-c': ,.,.\ 11-c', wherein R1, R2, R9\ R9\ W, and=-= are as defined in connection with Formula 11-c.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent l of a glucocorticosteroid having Formula 11-d: ,.,.\ 11-d. wherein R1, R2, R9\ R9\ W, and=-= are as d in connection with Formula 11-c.
In another embodiment, disclosed herein is a compound having Formula I-a, wherein SM is a monovalent radical ofa glucocorticosteroid having Formula 11-d': ,,,.\ 11-d'. n R1, R2, R9\ R9\ W, and=-= are as defined in connection with Formula 11-c.
In another embodiment, disclosed herein is a compound having a I-a, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent l of a glucocorticosteroid having Formula 11-e: 11-e, wherein: R1, R2, W, and =-= are as defined in connection with Formula 11-c; R9c is selected from the group ting of hydrogen, C1 4 alkyl, and -C(=O)R9\ R9ct is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally tuted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; and R9e is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl.
In another embodiment, disclosed herein is a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-e': w,,,. 11-e', wherein R1, R2, W, R9C, R9ct, and=-= are as d in connection with Formula 11-e.
In another ment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent l of a glucocorticosteroid having Formula 11-f: 11-f, wherein: R1, R2, R9C, R9ct , W, and=-= are as defined in connection with Formula 11-e.
In another embodiment, sed herein is a nd having Formula I-a, wherein SM is a monovalent radical ofa orticosteroid having Formula 11-f': w.,,, 11-f', wherein R1, R2, R9C, R9ct , W, and=-= are as d in connection with Formula 11-e.
In another embodiment, disclosed herein is a compound having Formula I-b: (SM-L-Q)n-A2 I-b or a pharmac eutically acceptable salt thereof, wherein: ] A2 is a protein; L is a linker; Q is a heterobifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a monovalent radical of a glucocorticosteroid having any one of: (1) Formula 11-1: O R3�Jrz XxxY� _,, .,,o R9f I I s ' '::/- 6e o 6 / R R c ·•10 11b R R6d 11-1; (2) Formula 11-m: O R3 tJ Z X*Y� _..,I R91 I ,, "'0--J··'' �' e R 6 ,,::, R 6 c '"0 R/11b ct 11-m; ] (3) Formula 11-n: O R 3 z 1391 I X*Y� r>I ,,.. .,,o : /'' J e ,,:;, -P; R6 R6c '"0 R 11b ct R2 11-n; (4) Formula 11-o: 0 .. R' Z X R"° R91 1/ I I '-"::: * e y2 ,,. ,oJliJ ,,::, \ ,,,0 R11b d R2 11-o; (5) Formula 11-p: 0 R' Z R91 1/ I )¢I · " 0 '�J e '-"::: ,,::, R ( y2 ,,. \ '"0 11b R ct R2 11-p; or (6) Formula 11-q: 0 R ' Z X R� 13 91 ,... I I ---:::: -n_ * ,,. \ J e ,,::, y2 '10 R 6 ' R11b ct R2 11-q, wherein: R1 is selected from the group consisting of hydrogen and halo; ] R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group consisting of -CH2OH, -CH2SH, -CH2Cl, -SCH2Cl, -SCH2F, /....... _Q / �0.._ /? ...__,,, y 'l(O R3b /_, _Q R3c ...__,,, 'l( p I -o-R3d R3a O O ' and O-R3e R3a is selected from the group consisting of hydrogen an d C1 4 alkyl; R3b is selected from the group consisting of C1 4 alkyl an d C1 4 alkoxy; R3c is ed from the group consisting of hydrogen, C1 4 alkyl, -CH20H, an d C1 4 alkoxy; R3ct and R3e are independently selected from hydrogen an d Ci-4 alkyl; ] R6\ R6b, R6C, R6ct , and R6e are each ndently selected from the group consisting of hydrogen, halo, C1 4 alkyl, C1 4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, an d alkoxy; Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=0)-, -S(=0)r, -NR5-, - CH2S-, -CH20-, -N(H)C(R8a)(R8b)-, CR4ct - (including both E an d Z isomers), an d -C=C-; (wherein when X is -CH2S-, , or -N(H)C(R8a)(R8b)-, the hetereoatom of -CH2S-, -CH20-, or (R8a)(R8b)- can be attached to either 6-membered ring, i.e., -CH2S- is equivalent to -SCHr, -CH20- is equivalent to -0CHr, and -N(H)C(R8a)(R8b)- is equivalent to -C(R8a)(R8b)N(H)-); or Xis absent, i.e., Xrepresents a chemical bond; Y2 is selected from the group ting of , -S-, an d -N(R7a)-; or Y2 is absent, i.e., Y2 represents a chemical bond; t is 1 or 2; Z is selected from the group consisting of =CR11a- an d N-; ] each R4a an d R4b are independently selected from the group consisting of hydrogen an d Ci-4 alkyl; or R4a an d R4b taken together with the carbon atom to which they are ed form a 3- to 6-membered cycloalkyl; R4c an d R4ct are independently selected from the group consisting of hydrogen an d C1 4 alkyl; R5 is selected from the group consisting of hydrogen an d C1 4 alkyl; R7a is selected from the group consisting of hydrogen an d Ci-4 alkyl; R8a an d R8b are independently selected from the group consisting of hydrogen an d Ci-4 alkyl; R9f is selected from the group consisting of hydrogen and Ci-4 alkyl; R11a an d R11b are independently selected from the group consisting of en, halo, C1 4 alkyl, C1 4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, an d alkoxy; and ] =-= represents a single or double bond.
In an other ment, disclosed herein is a compound having Formula 1-b: (SM-L-Q)n-A2 1-b, wherein: A2 is a protein; L is a linker; Q is a heterobifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and ] SM is a monovalent radical having any one of: (1) Formula 11-1': O R3 Z X*Y� ..rl R9f I I ,.- 6e ✓,; R6c ;g, R '''0 R11b R6d 11-1'; (2) Formula 11-m': (3) Formula 11-n': (4) Formula 11-o': 0 R3 Z X � R6b f:s 9f r. Y � o 2,,. \ ''' 1R6 R11b e y 0 R6d R2 11-o'; (5) Formula 11-p': 0 R3 z X R6b ,.,o-}··r-y 6eR9f ,,,. *I � O y2,,.\ .,,0 � R R d 11-p'; or ] (6) Formula 11-q': 0 R 3 z X R 6b 13 9 ,.... I I --::: ,,,oW, ,,,::. e 2 ... \ R6 Y ' ' R 11b d 11-q', w ere1nh • RI R2 R 3, , , =-=-=, R6 a R 6c R6d R6e R9f, , , , , Rllb, y2 X, , and z are as d fie 1ned ·1n connection· with Formula 11-1.
In another embodiment, disclosed herein is a compound having Formula 1-b: (SM-L-Q)n-A2 1-b, wherein: ] A2 is a protein; L is a linker; Q is a heterobifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a monovalent radical having any one of: (1) a 11-1": O R 3 ,....
X*Y� _,., R9f ,,,o I 6e ,,,:;. R6c ·•1 w, R 0 11b R d 11-1"; ] (2) Formula 11-m": 0 R 3 N �X* R9f Y� _,., -J ,••' I 6 e ,o R 6 c ·•10 R R11b d _ R6 R2 11-m"; (3) Formula 11-n": O R 3 ,.... 13 9f I X*I Y� _,.,,.. ,o : 6e ,o c ''10 w, R R11b d R2 11-n"; (4) Formula 11-o": 0 :�:,&::\ - .,,0 R 11b R2 11-o"; (5) Formula 11-p": O R3 gfN X R6b .,,o--J..., I 6• I '° vz✓\ '''0 R11b R R6d 11-p"; or (6) Formula 11-q": O R3;9, X R6b 1391N I )¢c I ---::: "10 : 8 ✓ 6e 2 \ R Y '''0 R11b R6d 11-q "' with Formula 11-1, and the carbon atom marked with s either the er or the S-isomer when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with an"*" is the R-isomer. In another embodiment, the carbon atom marked with an"*"is the S-isomer.
In another embodiment, disclosed herein is a nd having Formula I-a or 1-b, or a pharmac eutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having a 11-1: R3 :rz X*Y�_,.
R9f s ' ·010 I I �- 0 6 p; /, R6e R c '''0 11b R R6d 11-1, wherein: R1 is selected from the group consisting ofhydrogen and halo; R2 is selected from the group ting ofhydrogen, halo, and hydroxy; R3 is selected from the group consisting of -CH2OH, -CH2SH, -CH2Cl, -SCH2Cl, -SCH2F, I...._ _Q /�o .... /? " y LJO R3b /....._" LJ_Q R3c �-o-R3d R3a O O ' and O-R3e R3a is selected from the group consisting ofhydrogen an d C i-4 alkyl; R3b is selected from the group consisting ofC1_4 alkyl an d C i-4 alkoxy; R3c is selected from the group consisting ofhydrogen, C1_4 alkyl, , an d C1_4 alkoxy; R3ct and R3e are independently selected from hydrogen an d Ci-4 alkyl; Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=0)-, -S(=0)r, -NR5-, - CH2S-, -CH20-, (R8a)(R8b)-, -CR4c=CR4ct-, an d -C=C-; or Xis absent; tis I or 2; Z is selected from the group ting of=CR11a- an d =N-; each R4a an d R4b are independently selected from the group consisting of hydrogen an d Ct-4 alkyl; or R4a an d R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c an d R4ct are independently selected from the group consisting ofhydrogen an d C 1 4 alkyl; R5 is selected from the group ting ofhydrogen an d C1_4 alkyl; ] R6\ R6C, R6ct, an d R6e are each independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, an d alkoxy; Y2 is selected from the group consisting of, -S-, an d -N(R7a)-; or Y2 is absent; R7a is selected from the group consisting ofhydrogen an d C i-4 alkyl; R8a an d R8b are independently selected from the group ting ofhydrogen an d C i-4 alkyl; R9f is selected from the group consisting ofhydrogen and C i-4 alkyl; R11a an d R11b are independently selected from the group ting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, y, thiol, amino, alkylthio, an d alkoxy; and =-= represents a single or double bond.
In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a orticosteroid having Formula 11-m: R3 Z X*y2 ,.... ,,,o--r,,C.J 6eR9f I � '-/ R .0 R6c '''0 R/11b R6d 11-m. w ere1nh • RI R2 R3, , , =-=-=, R6 a R 6c R 6d R6e R9f, , , , , Rllb, y2 X, , and z are as d fie 1ned ·1n connection· with Formula 11-1.
In another embodiment, disclosed herein is a compound having a I-a or lb, or a ceutically acceptable salt thereof, n SM is a monovalent radical of a glucocorticosteroid having Formula 11-n: R3 Jz X*Y�_,., 13 e1 I I s ' ,, ,o : �.... 6e .o R6c '''0--r; 1 R R11b R6d 11-n. w ere1nh • RI R2 R3, , , =-=-=, R6 a R 6c R6d R6e R9f, , , , , Rllb, y2 X, , and z are as d fie 1ned ·1n connection· with Formula 11-1.
In another embodiment, disclosed herein is a compound having a I-a or 1-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-o: R3 Z X R6b ,,,o;.,i;.J 6eR9f *I � R .0 y2.,, \ '''0 R11b 11-o, wherein R1, R2, R3, =-=-=, R6\ R6ct, R6\ R9f, R11\ Y2, X, and Z are as defined in connection with Formula 11-1; and R6b is selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and .
] In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-p: R3 Z X R6b ,..... .,,o_L,t1JR9f I *I '-:::: 7 6e o y2/ \ '''0 R11b R 11-p. . . . ] w ere1nh RI R2 R3, , , =-=-=, R6 a R6b R6d R6e R9f Rllb y2 X, , , , , , , , and Z are as d fie 1ned 1n connection with Formula 11-o.
In another ment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a orticosteroid having a 11-q: R3 Z X R6b ,,,o�J 6e9f13 I *I ---:::: o y2/\ '''0 R�;b R6d 11-q.
. RI R2 R 3 . . w ere1nh , , , =-=-=, R6 a R6b R6d R6e R9f Rllb, , , , , , y2 X, , and Z are as d fie 1ned 1n connection with Formula 11-o.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein=-=-= represents a double bond.
In another embodiment, sed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, n SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein R1 is selected from the group consisting ofhydrogen and fluoro.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, n SM is a monovalent l of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein R2 is selected from the group consisting ofhydrogen and fluoro.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt f, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b' 11-1' 11-m' 11-n' 11-o' 11-p' 11-q' 11-1' 11-m" 11-n" 11-o" 11-p" or 11-q" wherein R3 is' ' ' ' ' ' ' ' ' ' ' ' ' selected from the group consisting of -CH20H, -CH2Cl, -SCH2Cl, -SCH2F, and -OH.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1', 11-m", 11-n", 11-o", 11-p", or 11-q",wherein: R3 is selected from the group consisting of: /�o : OH 0 ·, l�o O R3b /�o R3C q, Y n n C02H R3a O O , and R3a is selected from the group consisting of hydrogen and ; R3b is selected from the group consisting of methyl, ethyl, isopropyl, isobutyl, y, ethoxy, isopropoxy, and isobutoxy; R3c is ed from the group consisting of hydrogen, , ethyl, -CH20H, methoxy, ethoxy, and poxy; R3ct and R3e are independently selected from the group consisting of hydrogen, methyl, and ethyl.
In another embodiment, disclosed herein is a nd having Formula I-a or 1-b, or a ceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of ae II-a, 11-b, 11-c, 11-d, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein R5 and R8a are independently ed from the group consisting of hydrogen and methyl.
In another ment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein Z is =CH-.
In another embodiment, disclosed herein is a compound having a I-a or 1-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein Z is =N-.
In another ment, disclosed herein is a nd having Formula I-a or I-b, or a pharmaceutically acceptable salt f, wherein SM is a lent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein R6\ R6ct, and R6e are hydrogen.
] In another embodiment, disclosed herein is a nd having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein Y2 is -N(R7a)-. In another embodiment, R7a is selected from the group consisting of hydrogen and methyl. In another embodiment, R7a is hydrogen. In another embodiment, R7a is methyl.
In r embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", n: X is selected from the group consisting of R4b)t-, , -S-, -S( 0)-, -S( 0)r, -CH2S-, and -N(H)CH(R8a)-; tis l; R4a and R4b are independently selected from the group consisting of hydrogen and methyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- membered cycloalkyl; and R8a is selected from the group consisting of hydrogen and methyl. In another embodiment, X is -CH2-. In another embodiment, Xis selected from the group consisting of: In r embodiment, Xis . In r embodiment, Xis -S-. In another embodiment, Xis -CH2S-. In another embodiment, Xis -N(H)CHr. In another embodiment, Xis selected from the group consisting of: ] In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-1, 11-m, or 11-n, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-1', 11-m', 11-n', 11-1", 11-m", or 11-n", wherein R6c is selected from the group consisting of hydrogen, -Cl, -OMe (or -OCH3), and -OH.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt f, wherein SM is a monovalent l of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-o', 11-p', 11-q', 11-o", 11-p", or 11-q" n R6b is selected from the group consisting of hydrogen, -Cl, -OMe (or -OCH3), and -OH.
In another embodiment, disclosed herein is a compound having a I-a or I-b, or a pharmaceutically acceptable salt thereof, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein R9f is hydrogen.
In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a ceutically acceptable salt thereof, wherein SM is a monovalent l of a glucocorticosteroid having any one of Formulae11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein R9f is methyl.
In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt f, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae 11-1', 11-m', 11-n', 11-o'' ' ' ' ' ' ' ' '11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein R11a is selected from the group consisting of hydrogen and -OH.
In r embodiment, disclosed herein is a compound having a I-a or 1-b, or a pharmaceutically acceptable salt thereof, n SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein R11h is hydrogen.
In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt f, e.g. a compound having a I-a or 1-b wherein SM is a lent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein Lis a cleavable . In another embodiment, the cleavable linker comprises a succinimide, amide, thiourea, thioether, oxime, or self-immolative group, or a combination f. In another embodiment, the cleavable linker comprises a peptide. In another embodiment, the cleavable linker comprises a tripeptide. In another embodiment, the cleavable linker comprises a dipeptide. In another embodiment, the cleavable linker comprises phosphate ester. In another embodiment, the cleavable linker comprises a pyrophosphate diester.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, e.g. a nd having Formula I-a or I-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein Q is absent.
In another ment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or I-b n SM is a lent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein Q is a heterobifunctional group.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or I-b wherein SM is a lent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of ae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein Q is a bifunctional group selected from the group consisting of: ;�s-l H00C y-�JI H .._ N s_,,..._ Q-3 Q-4 Q-1 Q-2 /...,_ o s-\ \�� {J() .,J,., wherein m is 1, 2, 3, 4, 5, or 6. In another embodiment, Q is selected from the group consisting of Q-1, Q-2, Q-3, and Q-4. In another embodiment, Q is selected from the group consisting of Q-3 and Q-4. In another embodiment, mis 2.
In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt thereof, e.g. a nd having Formula I-a or 1-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of ae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein Q is a heterotrifunctional group.
In r embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or 1-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n' 11-o' 11-p' 11-q' 11-1"' ' ' ' ' ' ' ' ' ' 11-m" 11-n" 11-o" 11-p" or 11-q" wherein Q is a heterotrifunctional group that is: In another ment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or I-b wherein SM is a lent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n' 11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" n -L-Q- is·' ' ' ' ' ' ' ' ' ' .
LQ-1; m is 2 or 3; and R10a and R10b are independently selected from the group consisting ofhydrogen and optionally substituted C16 alkyl. In another ment, m is 2. In another embodiment, m is 1. In another embodiment, -L-Q-is: \5y�;:�yo s-� R10a O H O LQ-2.
In another embodiment, -L-Q-is: o s-� \ =��- XOb � � If N _N _,.. � '{ R10a O H O LQ-3.
In another embodiment, -L-Q-is: \5y�;:��o s-� R1oa O H O LQ-4.
In r embodiment, -L-Q-is: o s-� � � � \ � ll _.. �N'{ R10a O H O LQ-5.
In another embodiment, disclosed herein is a nd having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or I-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of ae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is·. m is 2 or 3; and R10a and R10b are independently selected from the group consisting ofhydrogen and optionally substituted C1_6 alkyl. In another ment, mis 2. In another embodiment, -L-Q- is: HO C 0 1310b 0 H H )y N N \ �N� S/ R1oa O H O LQ-7.
In another embodiment, -L-Q- is: HO C 0 R10b 0 ��� � ,.. ll � s/\ \ : IfR. _N C"'7m 1oa O H O LQ-8.
In another embodiment, -L-Q- is: HO C 0 R10b O H H \)yNY' �N S/ R1oa O NH O LQ-9.
In another embodiment, -L-Q- is: HO C 0 H �10b 0 H �N� ) \ ,..ll N \ : II 8✓ R.1o HN C"'7m a O O LQ-10.
] In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically able salt thereof, e.g. a compound having Formula I-a or I-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", n Lis a noncleavable linker. In another embodiment, the linker comprises one or more polyethylene glycol units.
] In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having a I-a or I-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is·.
LQ-11; m is 2 or 3; and xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15. In another ment, mis 2.
In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or 1-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is·. 0 s-i a a h � f-._ Q) /'-.. il NH C02H \ 0-\......,,, r _N/� X H LQ-12; m is 2 or 3; and xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15. In another embodiment, mis 2.
In another embodiment, disclosed herein is a nd having Formula I-a or 1-b, or a pharmaceutically able salt thereof, e.g. a compound having Formula I-a or 1-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is·. 0 0 \�O� Y.'P-\ 0 / LQ-13; and xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15.
The compound of any one of claims 1-47, or a pharmaceutically acceptable salt or e thereof, wherein -L-Q-is: 0 0 \ �o�0J:'�(s C02H LQ-29; and xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceu tically acceptable salt thereof, e.g. a compound having Formula I-a or I-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is·. mis 1 or 2; x is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; and R10a and R10b are independently selected from the group consisting of hydrogen and optionally tuted C 1.6 alkyl. In r embodiment, -L-Q-is: In r embodiment, -L-Q- is: In another embodiment, -L-Q- is: In another embodiment, -L-Q- is: In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceu tically acceptable salt thereof, e.g. a compound having Formula I-a or 1-b n SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is·. o s-l 0 R10b O O O NH \ )y�0N�O� rN¾i C02H R10a O H X H LQ-19; mis 1 or 2; x is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; and R10a and R10b are independently selected fromthe group consisting of hydrogen and optionally substituted C1.6 alkyl.
In another embodiment, -L-Q- is: In another embodiment, -L-Q- is: In another ment, -L-Q- is: ] In another embodiment, -L-Q- is: ] In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or 1-b wherein SM is a monovalent l of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of ae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n' 11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is·' ' ' ' ' ' ' ' ' ' . 0 R10b O 0 \��r��O� =>-S :,, LQ-24; xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; and R10a and R10b are ndently selected from the group consisting ofhydrogen and optionally substituted C1_6 alkyl.
In another embodiment, -L-Q- is: In another embodiment, -L-Q- is: 0 R10b O O :'(' �O�OY. \�, � ):1>- \ o / LQ-27.
In another ment, -L-Q- is: 0 R10b O 0 \)y�Y' N�O� rNJ( S R10a O X }-r \ o / LQ-28.
In another embodiment, disclosed herein is a compound having Formula I-a or 1-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or 1-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n' 11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is·' ' ' ' ' ' ' ' ' ' . 0 R10b O 0 \�:r�A/-- O� �(s C02H LQ-30; xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; and R10a and R10b are independently selected from the group consisting of hydrogen and optionally substituted C1_6 alkyl.
In another embodiment, -L-Q- is: ] In another embodiment, -L-Q- is: In another ment, -L-Q- is: \�:r��of----0 R10b O 01;" �(\,0 C02H LQ-33.
In another embodiment, -L-Q- is: 0 R10b O O \)y�0N� or��S of---- R10a O H x C02H LQ-34.
In another embodiment, disclosed herein is a nd having Formula I-a or 1-b, or a pharmaceu tically acceptable salt thereof, e.g. a compound having Formula I-a or 1-b wherein SM is a lent radical of a glucocortico steroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n'' ' ' ' ' ' ' ' ' '11-o' 11-p' 11-q' 11-1" 11-m" 11-n" 11-o" 11-p" or 11-q" wherein -L-Q- is any one of the chemical structures of Table I: Table I I�0� H H ,,,,,.....,___,o� ,,,,,.....,___, N N � :\, 0 0 � 8 ./ o O 0 ' ('o J o ) ol (' o H 02C I o o ( ( of o l__� s ./ "\. / � lf o o__) l___o o O ' H 02C / o� ./"-...../ o� o� �s./\ � o o ./"-...../ o ....,___,� 0 0 /' H HL-1 Y"'--' o� 'l. o� o� o� 0� 0� 0� 0 �N N� ,,,,.,. 0 Y"--' 0 0 ' I� o� o,,,,,.....,___, o� o �����s./"\ 0 0 0 H02CI /� o�o� o�o�� s ,,,,. '\., 0 0 ' In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, e.g. a compound having Formula I-a or I-b wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein n is 2-8. In another embodiment, n is 1-5. In another embodiment, n is 2-5. In another ment, n is 1. In r ment, n 1s 2. In another embodiment n is 3. In another embodiment, n is 4. In another embodiment, n 1s 5. In another embodiment, n is 6. In another ment, n is 7. In another embodiment, n is 8.
In another embodiment, disclosed herein is a compound having Formula I-a or I-b, or a pharmaceutically acceptable salt thereof, wherein SM is a lent radical of a glucocorticosteroid which is any one ofthe chemical structures of Table II.
Table II o �s ... ,o� h .,,o "=<N­\ F F 0 s lo ..,,� I I 'QO ·" N,....\ .,,o7 H OH su OH vs 0 0 H .. 0 ..,, 0- N ,,o N ,....\ .,,o H ··" 7 ' •HQ - .,.,.,, F F OH S ...,a s � 7 ,,,0- ...,o b-�, ,,,o p ,,,o ;r" 0 0 F .,,,.. F .. ,,07s ,,, o p 0 HN i= i= .. ,a 9'6 ::::,... 1 ,....\ F F o · ....o�r9'1 �s :::,,. ,....\ i= i= .. ,o � ;; 0 NH O OH H '/ 0 O , , , ...,07" .. ,o "0- - p� 0 ' F ,,.,.. �' 0 HO / ..o-r(}- '0 .. ,o � h ..,0 y 0 i= OH l" vec"-:::I I NH ,,,, ,e:;:: A ..,,07 Cl OH I I � NH �,e:;:: A .,110 Cl 0 OH OH ···,,� .. ,o I i= ' 1/' I ::::.... ... ,o� .,, o I F � I '/ ' · '/ ' F 0 -- -10 JJ �--} (S ,.... 07 HN-/ H��J_cr-q / H oO- HO / OH HO 0 2fv 0 ,•' .. ,o - .. ,0 o OH .J)�s .. ,,07· ·••0 Q, � ,..-\ 10471 WO OH S � 0 V""' U""' 1 ...,07· ,,, ,0 ,0 Cl OH s' OH S a ·•''V� . N� 0 N ,,.-\ � �N/\ "'' 7·••O ..110� .,,o ,, H u ��-j'-ff OH r('Y S� ...,o--,;.:•'V.,,o � �N,.., \ and OH In r embodiment, disclosed herein is a compound having a I-a or I-b, or a pharmaceutically able salt thereof, n SM is a monovalent radical of a glucocorticosteroid selected from the group consisting of: 1�1/"'-..J _✓,; 0 "'.'.d"�I C 'o l � "''.�J••,,�""'o ::,...
.,,- N'/· and' In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt f, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of ae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 is an antibody or antigen-binding fragment thereof or wherein A2 is an antibody or antigenbinding fragment thereof.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent l of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having a 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 is an anti-tumor necrosis factor (TNF) alpha protein that binds to human TNF alpha and/or murine TNF alpha or wherein A2 is protein that binds to human TNF alpha and/or murine TNF alpha.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a nd having Formula I-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one ofFormulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 is an anti-tumor necrosis factor (TNF) alpha n that binds to soluble TNF alpha or n A2 is protein that binds to soluble TNF alpha.
In r embodiment, disclosed herein 1s a compound having Formula I-a, or a pharmaceutically able salt f, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula I-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-b, wherein SM is a monovalent radical of a orticosteroid having any one ulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 is an anti-tumor necrosis factor (TNF) alpha protein that binds to membrane-bound TNF alpha or wherein A2 is a protein that binds to membrane-bound TNF alpha.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having a I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula I-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one ofFormulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 is an anti-tumor is factor (TNF) alpha protein comprising an anti-TNF antibody or wherein A2 is protein comprising an anti-TNF antibody.
In another embodiment, disclosed herein 1s a compound having a I-a, or a pharmaceutically able salt thereof, e.g., a nd having Formula I-a, wherein SM is a lent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically able salt thereof, e.g., a compound having Formula 1-b, wherein SM is a lent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 is an anti-tumor necrosis factor (TNF) alpha protein comprising an anti-TNF receptor antibody or wherein A2 is a protein comprising an anti-TNF receptor antibody.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent l of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having a 1-b, or a pharmaceutically acceptable salt thereof, e.g., a nd having Formula 1-b, n SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 is an anti-tumor necrosis factor (TNF) alpha protein comprising an n-binding nt of an anti-TNF antibody or wherein A2 is a n comprising an antigen-binding fragment of an anti­ TNF antibody.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one ofFormulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 is an anti-tumor necrosis factor (TNF) alpha protein comprising an antigen-binding fragment of an anti-TNF receptor antibody or wherein A2 is an umor necrosis factor (TNF) alpha protein comprising an antigen-binding fragment ofan anti-TNF receptor antibody.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt f, e.g., a compound having a I-a, n SM is a monovalent radical of a orticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11- q",wherein the n-binding fragment is selected from the group consisting of Fab, Fab', F(ab')2, single chain Fv or scFv, disulfide linked Fv, V-NAR domain, IgNar, ody, IgGACH2, minibody, F(ab')3, tetrabody, triabody, diabody, single-domain antibody, DVD-lg, Fcab, mAb2, (scFv)2, or scFv-Fc.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein the antibody or antigen-binding fragment thereof is murine, chimeric, zed, or human.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having a I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having a 1-b, or a pharmaceutically acceptable salt f, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11- rein A1 is an umor necrosis factor (TNF) alpha protein comprising a e TNF or or wherein A2 is a protein comprising a soluble TNF receptor. In another embodiment, the soluble TNF or is a soluble p75 TNF receptor.
In another embodiment, disclosed herein is a compound having Formula I-a, or a ceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having a 1-b, or a pharmaceutically able salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of ae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one ofFormulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 comprises a heavy chain constant domain or a fragment thereof or wherein or A2 comprises a heavy chain constant domain or a fragment thereof. In another embodiment, the heavy chain constant domain or nt thereof comprises a constant domain selected from the group consisting of:(a) an IgA nt ; (b) an IgD constant domain; ( c) an IgE constant domain; ( d) an IgG 1 constant domain;(e) an IgG2 constant domain; (f) an IgG3 constant domain; (g) an IgG4 constant domain; and (h) an IgM nt domain or is a nt thereof. In another embodiment, the heavy chain constant domain comprises a human IgG 1 heavy chain constant domain or fragment thereof. In another embodiment, the heavy chain constant domain comprises a human IgG1 Fe domain.
In another ment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically able salt thereof, e.g., a nd having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, wherein A1 comprises a light chain constant domain or a fragment thereof or wherein A2 comprises a light chain constant domain or a fragment thereof. In r embodiment, the light chain constant domain or fragment thereof comprises a nt domain selected group consisting of (a) an lg kappa constant domain and (b) an lg lambda constant domain or is a nt thereof.
In another embodiment, disclosed herein is a compound having a I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of ae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a orticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1competitively inhibits binding of an antibody ed from the group consisting of adalimumab, infliximab, certolizumab pegol, and golimumab to TNF-alpha or wherein A2 competitively inhibits binding of an antibody selected from the group consisting of adalimumab, infliximab, certolizumab pegol, and golimumab to TNF-alpha.
In another embodiment, disclosed herein 1s a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of ae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1binds to the same TNF-alpha epitope as an antibody selected from the group consisting of adalimumab, imab, certolizumab pegol, afelimomab, nerelimomab, izumab, placulumab, and golimumab or wherein A2 binds to the same TNF-alpha epitope as an antibody ed from the group consisting of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having a I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having a I-b, or a pharmaceutically able salt thereof, e.g., a compound having Formula I-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein the anti-TNF alpha protein is selected from the group consisting of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab.
In another embodiment, disclosed herein is a nd having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a lent l of a orticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 comprises the variable heavy chain CDRl, CDR2, and CDR3 ces of SEQ ID N0:3 or 6, SEQ ID N0:4, and SEQ ID N0:5, respectively and the variable light chain CDRl, CDR2, and CDR3 ces of SEQ ID N0:32, SEQ ID N0:3 3, and SEQ ID N0:3 4, respectively or wherein A2 comprises the variable heavy chain CDRl, CDR2, and CDR3 sequences of SEQ ID N0:3 or 6, SEQ ID N0:4, and SEQ ID N0:5 tively and the variable light chain CDRl, CDR2, and CDR3 sequences of SEQ ID N0:32, SEQ ID N0:3 3, and SEQ ID N0:34, respectively.
In another embodiment, disclosed herein 1s a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a nd having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, n SM is a lent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 comprises the variable heavy chain sequence of SEQ ID NO:50 and the variable light chain sequence of SEQ ID NO:59 or wherein A2 comprises the variable heavy chain sequence of SEQ ID NO:50 and the variable light chain sequence of SEQ ID NO:59.
In another embodiment, disclosed herein 1s a compound having Formula I-a, or a ceutically acceptable salt thereof, e.g., a compound having a I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a nd having Formula 1-b, or a pharmaceutically acceptable salt f, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1does not bind to TNF beta or wherein A2 does not bind to TNF beta.
In another embodiment, sed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a nd having Formula 1-b, or a pharmaceutically acceptable salt f, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of ae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 binds to TNF beta or wherein A1 binds to TNF beta.
] In r embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one ofFormulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one ofFormulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1neutralizes human TNF-alpha cytotoxicity in a in vitro L929 assay with an IC50 of lXl0 7 M or less or wherein A2 neutralizes human TNF-alpha cytotoxicity in a in vitro L929 assay with an IC50 of 1x10 7 Mor less.
In another embodiment, disclosed herein is a compound having a I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having a I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula I-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one ofFormulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 blocks the interaction of TNF-alpha with p55 and p75 cell surface receptors or wherein A2 blocks the interaction of TNF-alpha with p55 and p75 cell surface receptors.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-a, n SM is a monovalent radical of a orticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula I-b, or a pharmaceutically acceptable salt thereof, e.g., a nd having Formula I-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one ofFormulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein A1 lyses surface TNF expressing cells in vitro in the presence of complement or n A2 lyses surface TNF expressing cells in vitro in the presence of complement.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, e.g., a compound having a I-a, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula I-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula I-b, wherein SM is a monovalent radical of a orticosteroid having any one of ae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one ulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein the soluble p75 TNF receptor is etanercept.
In another embodiment, disclosed herein 1s a compound having Formula I-a, or a pharmaceutically acceptable salt f, e.g., a compound having Formula I-a, n SM is a monovalent radical of a glucocorticosteroid having any one of Formulae II-a, 11-b, 11-c, 11-d, 11-e, 11-f, 11-1, 11-m, 11-n, 11-o, 11-p, or 11-q, or any one of Formulae II-a', 11-b', 11-c', 11-d', 11-e', 11-f', 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, e.g., a compound having Formula 1-b, wherein SM is a monovalent radical of a glucocorticosteroid having any one of Formulae 11-1, 11-m, 11-n, 11-o, 11-p, or 11- q, or any one of Formulae 11-1', 11-m', 11-n', 11-o', 11-p', 11-q', 11-1", 11-m", 11-n", 11-o", 11-p", or 11-q", wherein the antibody is adalimumab.
In another embodiment, disclosed herein is a nd having Formula I-a, or a pharmaceutically acceptable salt thereof, or a nd having Formula 1-b, or a pharmaceutically acceptable salt thereof, which is any one ofthe al structures of Table III: Table III H02 C 0 O H o ° - 0 ��� � s _ � A H '. )l_ N A H-/- N'('-' s HO N �)l,, ,0 l N !f°H 1 0 ° ---( H O H02C 0 o 0 HO O o )l.,:- N H02 C _ 0 H H � H ' )l_ N N'('-' s A N !f°H 1 0 ° HO O 0 --i H02 C � )--/� H 11 s A H H _ O H 0 H \ )l__,,N N� S A H O l 'l{'-' 0 0 ,,,UV ' O 0 HO O Ny"�0 ,· --i ,,,10 0 ' 0 � 0 0 HO C o s A HO O 0 � i,,,.,__ N ,ll_,,, � � S A � H 0 V-- J\ H O - ,.,.......___,, 0-.....,/""'-- ,,,,.......__,, � H H O 00� _) N� _, 0 O 0 � O 0 � o--,.••'v 0 - H .,,0 0 0 o o s A H A../'- C HO O � N,ll_,,, � � N H � 1 · · ,,V �N H H � ) N S A H O HO2C H O � ,...__,,O-../"o ,,� N� _, 0 O 0 0 HO O 0 - H o .••'OV s A 0 0 HO o � , () ( A 1 · · '(\)l_,, �½' N� H \ ✓� � o Ho,c )l__,� v�----- : HO O ,,,,U S D-- � \ -; H 0 0 H�2C H 0 0 H A D-- 0 s A N,ll.__,, )l__, �-;-N � s H ) HO O , .. ,V ('>r 0 H�,c s H0 2 C o s A 0 H H o = H � o ,,V �_J__)l,,-)l, 0 N S A v--N H H _ ) ! H ' )-._,, N II N � . ._( H O HO 2 C \ 0 O 0 HO l(' � o � � H -i·· )l.) H .,,0 = 0 H H � H S )___ '("" N S ., N A O l" �- \ HO � 0 O O �OH H02C ' S A '117- �!'(}1 <O ;v-,� so o O l) �OH ·· H \ 0 C H0 2 o.._( ,10 ,10 . - 0 H �S H0 N A 0--.,/'- ,,.....__,., N-.../"-' H O \\ O N 0 ¢ s A --i("' � o � .,,)l.._r -N� HO O 0 O H V HO .••' l) H ) a-- O , ,, N� 1· � H O C H02 , .,10 - \ \_ H�,c s A � l__ r�--r- 0 _ � H N y-- A "' N�0 --ir-- N 0 H ' s N H \ ,osvNl� � O H 0 � 0 O O HO HO .• .,, /2 N O 0 .,,0 .,,O --, - - 0 0 H02C1l' 1{ll1\ \_ A <0 0 ) V o, ..l = 0 H )l___, N}S A (' Ho ,-; � ' NY"' (' o) o J o1 o 0 o o � N o l....- � s A ()Vi "" - ( y- \ O o -H ( o y"-' ( J 0 ,4, 0 HO O o o__..) l..,,., o o HO O \ � # o-- !l)' 0 ,,, ( ,,10 ,· A o) 0 (' o "�{'c H (' o J 1 �"y"-' o ( ) o o l..,,.,N s A o o ( J HO 0 0 )!.._ ) o l.._.,6 o__..) H0 2 C _ 0 H A ! H N � S � A O O HO O --,, 'i("H --,, 0 O _ •• ,UV H02 C - 0 H H H ' � N � S A s--,('y --('� \ � o HO O V 0 0 ,· •'O .. ,o HO,}C � 0 H S S A jl,,__ N� N S A N N O�o � S )!J-..
H _,---. �O�o � O Q H 0 O H N� � N� � Y' HO H OC 2 0--. ( ' v � O � (YO / 0 o ] · ,10 ) 0 /2' 0tJBt 0 . ••' UC t° ' - A , 0 0 .. ,o !_..-• A CY � , )l____,--... N \1N �S H H 0 H0 2C ) H0 2 C H � S N N S A A O ('Y O 0 HO O 0--(-v 1-{ N:1 '" ,� A s A 0 �) � HO O � ,Ni 0 ,.�Sf; ,0 01)l 1 � ,10 Y\-- 0 ' 0 . r:; '-O �p'0 s ,o O o__r' .v s A HO o d'0-,)l ) �N \\ H ) H \ O C 0 . "" ,do-}ff�l; H O C C ' I = H0 2 � /"" �� s A �}s A ""o/"-.,/ o./-- o �p1H \ o � � � NL�--(""' 0 -- �✓O-./ 0 � /"-.,/ o ) 0 s HO o (" --Y 'K ° � _f�f0 o )L__� HO O __ o Ql .,O ( . .,,0 if /·. i ' - \ --i('-" ''_,.,. 0--../'- O.__./'- Q' � ) H 02C HO O OS� V 0 HN o 0 o s A §) � .., N�N� A H �N.._,,.......... � H N H _) O H02C O -,· H HO o V 0 ' wherein n is 1-5 and A is A1 or A2. In another embodiment, A is adalimumab.
In another embodiment, disclosed herein is a nd having Formula I-a, or a pharmaceutically acceptable salt thereof, or a compound having Formula 1-b, or a pharmaceutically acceptable salt thereof, which is any one of the chemical ures ofTable IV: n 2 2 4 s+A + • A , 0 N� H _ ) 00� o HO,C \ 2 C 0 O Ht\ 0 NY' o I H N \ 0 i-'y' H � o C l_ 0 ! I ? 0- - v"' l.__,,.o ( \ H N 0 Structure bl O c\L � 0 l_ ) 0 h '('N O] o \ .0 07 � o -0 t___,o \) l)' H --:: N 0 0 A/ -- r/ J; O '<"i' V-- � A/'- V--� ("Y •'v 0 1c - y o H 0 '?\1 ,,,V O ,0 O HO � ' F HO ,0 � i' 0 0� o �1 - - 96 Table IV n 4.5 2 4.4 A n A n n A S 0 ;l_,., s ) 0 - N H0 2C - N � H H02C H_) H02C ?, ,.,,..___,,.,., � H ?i ,.,,..___,,.,., � l/'N�s H ' . N 0 = , 0 , / HN �� j__ .,,'' �H HN � Structure o �-H "0'-- 1 0' 1 o, a' 1 o � H �N/ 0 , , , I .•,0 -.,. ('Y ,,V�N · ' 0 o � I _.,O - O - o - :. F HO :. F i" ,0 ,0 , o Ho 0 � 0 * 0 0 0 0 0 0 � � � <( C poJ:ZI ,1 IU.. 0 0 0 0 � � CZ� ',,'" ••ILL � oJ:"�s� ..... ( .... -<; o ,,_ r ,, __ / I rI 0 / I z � o�� Q,,_ 0 _ 0 ,,__10 0 0 0 0 •11LL .. ,u.. 0 0 -mofi- 0 0 0 'I ILL 0 0 :;; 0 "'d ("") 00 N 0 '""' (,,; Ut Ut '""' QC) N 0 '""' ---l -- N '""' 0 ---l '""' 3.8 a compound A n thereof, or S I I O N H�) H N 'I(' � O ceutically acceptable salt Table IV-A: S'--_/-::::::,../ structures of .,,0 F or a �,, v ::7 , the chemical 0'1/ I I \ - I Formula I-a, one of - 105 is any I 2 4 compound having Table IV-A A n A S " _ ) 0 C _, s is a thereof, which 6 H02 H02 C " is adalimumab. O ·,, � I lr) l HN� H N t) o y' ,) " I o 0 H� acceptable salt � . N H N i\" J\ (', U � 'CJ o embodiment, A disclosed herein � � !, --V I ,,\0 o� .. I ,,\0 In another ment, a pharmaceutically 2 . o o " HO �� - - uO � 1 or A another In I-b, or 0 wherein A is A [00462] having Formula Io,�o wherein A is A1 or A2. In r embodiment, A is adalimumab.
In another ment, disclosed herein is a compound having Formula I-a, or a ceutically acceptable salt thereof, or a compound having Formula I-b, or a pharmaceutically able salt thereof, which is any one ofthe chemical structures of Table V: Table V \ �N--< � � .� � V V,,,.(o H 0 ;---J NH 0 A sJN �H HO 0 0 0 I H O � 0'' 0 A s-{��f-'l(yl�� OH wherein n is 1-5 and A is A1 or A2. In another embodiment, A is adalimumab.
In another embodiment, disclosed herein is a compound having Formula I-a, or a pharmaceutically acceptable salt thereof, or a compound having Formula I-b, or a pharmaceutically acceptable salt thereof, selected from the group consisting of: l �1·•'t ·�- A s J--NJN , N� H H ,., r( O = H OH � C0 2H 0 0 l H O r-7) 0''' 0 A S-c��r1tl'�� wherein n is 2-4, A is A1 or A2. In another embodiment, A is adalimumab. In another embodiment, n is 2 or 4. In another embodiment, n is 2. In another embodiment, n is 4.
IV. orticoid receptor agonists In another embodiment, disclosed herein is a compound having Formula VII: or a ceutically acceptable salt thereof, wherein: R1 is ed from the group consisting ofhydrogen and halo; R2 is selected from the group ting ofhydrogen, halo, and hydroxy; R3 is selected from the group consisting of -CH20H, -CH2SH, -CH2Cl, -SCH2Cl, -SCH2F, R3a is selected from the group consisting ofhydrogen and Ci-4 alkyl; R3b is selected from the group consisting of C1_4 alkyl and C i-4 alkoxy; ] R3c is selected from the group consisting of hydrogen, C1_4 alkyl, -CH20H, C1_4 alkoxy, -CH2(amino), and -CH2CH2C(=O)OR3f; R3ct and R3e are independently selected from the group consisting ofhydrogen and Ci-4 alkyl; ] R3f is selected from the group consisting of hydrogen and C1_4 alkyl; Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=O)-, -S(=O)r, -NR5-, - CH2S-, -CH20-, -N(H)C(R8a)(R8b)-, -CR4c=CR4ct- (including both E and Z isomers), -C=C-, -N(R5)C(=O)-, and -OC(=O)-; (wherein when Xis -CH2S-, -CH20-, -N(H)C(R8a)(R8b)-, -N(R5)C(=O)-, or -OC(=O)-; the hetereoatom of -CH2S-, -CH2O-, -N(H)C(R8a)(R8b)-, -N(R5)C(=O)-, or -OC(=O)-; can be attached to either ered ring, i.e., -CH2S- is equivalent to -SCHr, -CH2O- is equivalent to -OCHr, -N(H)C(R8a)(R8b)-is equivalent to -C(R8a)(R8b)N(H)-), C(=O)- is equivalent to N(R5)C=O)­ and -OC( O)- is equivalent to -C( O)O-; or Xis absent, i.e., Xrepresents a chemical bond; ] tis 1 or 2; Z is selected from the group consisting of=CR11a- and N-; each R4a and R4b are independently selected from the group consisting of hydrogen and Ct-4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered lkyl; R4c and R4ct are ndently selected from the group consisting of hydrogen and Ci-4 alkyl; R5 is selected from the group consisting ofhydrogen and C1_4 alkyl; ] R6\ R6b, R6C, and R6ct are each independently selected from the group consisting of en, halo, C1_4 alkyl, haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; R7a is selected from the group consisting ofhydrogen and Ci-4 alkyl; R7b is selected from the group consisting of hydrogen, -L-H, -L-PG, 0 0 �� j L-N j L 0 , and O ; or R7a and R7b taken together with the nitrogen atom to which they are attached form: O ; or R7a and R7b taken together with the nitrogen atom to which they are attached form a nitro (-N02) group; mis 1, 2, 3, 4, 5, or 6; L is a linker; PG is a protecting group, e.g., Boe, FMOC; R9f is selected from the group consisting of hydrogen and Ci-4 alkyl; R8a and R8b are independently selected from the group consisting ogen and Ci-4 alkyl; R11a and R11b are independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, Cn haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond. In another embodiment, R7b is en. In another embodiment, R7b is ed from the group consisting of: 0 1310b Q H �10b \)y N���P \)y�0NH 2 R10a 0 R10a � , and mis 1, 2, 3, 4, 5, or 6; and R10a and R10b are each independently ed from the group consisting of hydrogen and optionally substituted C1-6 alkyl.
In another embodiment, disclosed herein is a compound having Formula VII': R6a 6b 0 R3 ,,. z x R R7b •IIQW DI N' 7a · •1 0 R11b 6c R6d R VII', or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9fR11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, disclosed herein is a compound having Formula VII": R6a 6b 0 R3 _,,. x R R7b RITT I DI N •110 ' 7a R ·•10 R11bp;r 6c R6d R VII", or a pharmaceutically acceptable salt thereof, n R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9fR11\ and X are as defined in tion with Formula VII, and the carbon atom marked with an"*"is either the R-isomer or the S-isomer when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with an"*"is the R-isomer. In another embodiment, the carbon atom marked with an"*"is the S-isomer.
In another embodiment, disclosed herein is a compound having FormulaVII-A or Formula VII-B: 0 R3 z x�6bl7b � r ,oyy -�N R7a . ·•10 R60 VII-A or R2 VII-B, or a pharmaceutically acceptable salt or solvate thereof, n: R1 is selected from the group consisting ofhydrogen and halo; R2 is ed from the group consisting ogen, halo, and y; R3 is selected from the group consisting of -CH20H, -CH2SH, -CH2Cl, -SCH2Cl, -SCH2F, - SCH2CF3, -CH20S( 0)20H, hydroxy, -OCH2CN, /'--.._..-,oyyoH ·,,0H R3a is selected from the group consisting ofhydrogen and Ci-4 alkyl; R3b is selected from the group consisting ofC1_4 alkyl and Ci-4 alkoxy; R3c is selected from the group consisting of hydrogen, C1_4 alkyl, -CH20H, C1_4 alkoxy, -CH- 2(amino), and -CH2CH2C( O)OR R3ct and R3e are independently selected from the group ting ofhydrogen and Ci-4 alkyl; R3f is selected from the group consisting of hydrogen and C1_4 alkyl;X is selected from the group consisting of -(CR4aR4b)t-, , -S-, -S( O)-, -S( O)r, -NR5-, -CH2S-, -CH20-, -N(H)C(R8a)(R8b)-, -CR4c CR4ct -, -C=C-, -N(R5)C( O)-, and -OC( O)-; or X is absent; tis 1 or 2; Z is selected from the group consisting of CR11a- and N-; each R4a and R4b are independently selected from the group consisting of hydrogen and Ci-4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group ting ofhydrogen and Ci-4 alkyl; R5 is selected from the group consisting ofhydrogen and C1_4 alkyl; R6\ R6b, and R6care each independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, haloalkyl, cyano, hydroxy, thiol, amino, hio, and ; R7a is selected from the group ting ofhydrogen and C1_4 alkyl; R7b is selected from the group ting ofhydrogen, -L-H, -L-PG, 0 , and j-L-N� 0 ; or R7a and R7b taken together with the nitrogen atom to which they are attached form: 0 ; or R7a and R7b taken together with the nitrogen atom to which they are attached form a nitro mis 1, 2, 3, 4, 5, or 6; L is a linker; PG is a protecting group; R9f is selected from the group consisting of hydrogen and C1 4 alkyl; ] R8a and R8b are independently selected from the group consisting ogen and Ci-4 alkyl; ] R11a and R11b are independently selected from the group consisting of hydrogen, halo, C1 4 alkyl, Cn haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond. In another embodiment, R7b is hydrogen. In another embodiment, R7b is selected from the group consisting of: 0 13 10b Q b H �10 \)y N���P \)y�0NH 2 R10a � R10a 0 , and mis 1, 2, 3, 4, 5, or 6; and R10a and R10b are each independently selected from the group consisting of hydrogen and optionally tuted C1-6 alkyl.
In another embodiment, sed herein 1s a compound having Formula VII-A' or Formula VII-B': 0 R3 Z XD R7bR6a o R3 �91 (z x� 7b WR I I N N ,11Q ' ?aR ..,o-yy/( _�N /R7a ·•10 R11b R6b R6c ·•10 R11b R60 0 0 VII-A' or VII-B', or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R7\ R7b, R9f, R11\ X, and Z are as definedin connection with Formula VII-A.
In another embodiment, disclosed herein 1s a nd having Formula VII-A" or Formula VII-B": 0 R6a R3 ,,.. X R7b 0R3 X R6a 6bR 9f I I N I I N R D R91 •IIQ DR7a '''0p;r ' ?aR ,,,o R6b R6c '''0p;} R11b R11b R6c 0 0 VII-A" or VII-B", or a pharma ceutically able salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R7\ R7b, R9f, R11\ and X, are as defined in connection with Formula VII-A, and the carbon atom marked with an "*" is either the er or the S-isomer when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with an "*" is the R-isomer. In another embodiment, the carbon atom marked with an "*" is the S-isomer.
In another embodiment, disclosed herein is a compound having Formula VIII: R3 :Jz x*: [ R9f I I R7a ·•• J:r; �t R6d ,,;:::; R6b ''' 11b 0 R 6c VIII, or a pharma ceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as d in connection with Formula VII.
In another embodiment, disclosed herein is a nd having Formula VIII': O R3 R9f I IZ X*: rR7a h 6b '''0 11b R6d R _ R R6c;g, R2 VIII', or a pharma ceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, disclosed herein is a compound having Formula VIII": R6a �7b 0R3 ,,.. X ---::: N, 9f I ◊I R7a .,,o�R 6d '''0 11bR ✓,:;. R6b R R6c VIII", or a pharma ally acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ and X are as defined in connection with Formula VII, and the carbon atom marked with an "*" is either the R-isomer or the S-isomer when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with an"*"is the R-isomer. In another embodiment, the carbon atom marked with an"*"is the S-isomer.
In another embodiment, disclosed herein is a compound having Formula VIII-a: R6a R?b R3 (Jz x*rt ?a R9f R ,,10�·•'' �-- I I 111b R6d ,0 R6b '''0 R R6c VIII-a, or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, sed herein is a compound having Formula ': R6a R7b o R3 �z x rtR7a R9f I *I ''10, 1,.,,/ R6d ✓,; R6b ''10 R11b R6c VIII-a', or a pharmaceutically able salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, disclosed herein is a compound having Formula VIII-a": R6a fb 0 N X '--::: N,R * 7a R9f 9, I I ''10' 1_.,,/ R6d ✓,; R6b ''10 R11b R6c VIII-a", or a pharmaceutically able salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ and X are as defined in connection with a VII, and the carbon atom marked with an"*"is either the R-isomer or the er when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with an"*"is the R-isomer. In another embodiment, the carbon atom marked with an"*"is the S-isomer.
In another embodiment, disclosed herein is a nd having Formula VIII-b: R3 Jz xx;c: [ 13 y-- R7a ,,10 : I I ,0 p; R6d R6b '''0 R11b R6c VIII-b, or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In r embodiment, disclosed herein is a compound having Formula VIII-b':*R6a 0 R3 ,,, z X N,R7a 13 9f HO I "'?,W'R• " R• R2 VIII-b', or a pharmaceutically able salt f, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, disclosed herein is a compound having a VIII-b": 6aR �7b 0 Rw,,,,3 X ,..,_ N, 9f "' laR 13 " I ;¢c I .,,o .-,:;;; 6R d 6R b '''0 R11b 6R c VIII-b", or a pharmaceutically able salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ and X are as defined in connection with Formula VII, and the carbon atom marked with an"*"is either the R-isomer or the er when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with an"*"is the R-isomer. In another embodiment, the carbon atom marked with an"*"is the S-isomer.
In another embodiment, disclosed herein is a compound having Formula IX: R3 Z X R6b ,,,o�J9 :;,- --.::::: I ,0 ,R7a R6d N R11b I '''0 R6c R7b or a pharmaceutically acceptable salt f, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, disclosed herein is a compound having Formula IX': 0 R3 z X R6b HO .,,o.Q ¢I : ,R7a _ .,,0 R11b R6d R6c J � I�, or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b, R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, disclosed herein is a compound having Formula IX": H ,:p;::(t1:>::,, .,, R6dy':'J 0 R11b _ R6c R7b R2 IX", or a pharma ceutically able salt thereof, wherein R1, R2, R3, =-=, R6\ R6b , R6C, R6ct , R7\ R7\ R9f, R11\ and X are as defined in connection with Formula VII, and the carbon atom marked with an"*"is either the R-isomer or the S-isomer when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with s the R-isomer. In another embodiment, the carbon atom marked with an"*"is the S-isomer.
In another ment, sed herein is a compound having Formula IX-a: IX-a, or a pharma ceutically able salt thereof, wherein R1, R2, R3, =-=, R6\ R6b , R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, disclosed herein is a compound having Formula IX-a': --t--rR3 Z X R6b 91 ,..- '--:::: d I ,0 'R7a '''0 �RR ' R6c R7b IX-a', or a pharma ceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b , R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, sed herein is a compound having Formula IX-a": R 3 x '--:::: R6b ,,,o-J--•'R91N I d I ,0 'R7a '''0 R11b R ' R6c R7b IX-a", or a pharma ceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b , R6C, R6ct , R7\ R7\ R9f, R11\ and X are as defined in connection with Formula VII, and the carbon atom marked with an"*"is either the R-isomer or the S-isomer when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with an"*"is the R-isomer. In another embodiment, the carbon atom marked with an"*"is the S-isomer.
In another embodiment, disclosed herein is a compound having Formula IX-b: IX-b. or a ceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b , R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in tion with Formula VII.
] In r embodiment, disclosed herein is a compound having Formula IX-b': 0 R3 f ,_..z X R6b HO ·"0 � / I .o ,R7 a - "' 0W, R"° " R" �'° R2 IX-b'. or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, =-=, R6\ R6b , R6C, R6ct , R7\ R7\ R9f, R11\ X, and Z are as defined in connection with Formula VII.
In another embodiment, disclosed herein is a compound having a IX-b": R3 R6b O X 91N �13 ¢ . ,,o : I I � R7a ,.;:;:; , .,,0 R11b t-;J R6c R7b IX-b". or a pharmaceutically acceptable salt f, wherein R1, R2, R3, =-=, R6\ R6b , R6C, R6ct , R7\ R7\ R9f, R11\ and X are as defined in connection with Formula VII, and the carbon atom marked with an"*"is either the R-isomer or the S-isomer when R2 is halo or hydroxyl. In one embodiment, the carbon atom marked with an"*"is the R-isomer. In another ment, the carbon atom marked with s the S-isomer.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', , VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein =-= represents a single or double bond.
In another embodiment, =-= represents a double bond.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R1 is selected from the group consisting of hydrogen and fluoro.
In another embodiment, disclosed herein is a compound having any one of ae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group consisting of hydrogen and fluoro.
In another embodiment, disclosed herein is a nd having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R3 is ed from the group consisting of -CH20H, -CH2Cl, -SCH2Cl, -SCH2F, and -OH.
In another ment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein: R3 is ed from the group consisting of: /'-._....,-0 ' OH 0 . /, Q O R 3b / ....._ _Q'y'R y, ''OH ,,, Y n " 11 COzH R3a O 0 R3a is selected from the group consisting of hydrogen and methyl; R3b is selected from the group consisting of methyl, ethyl, isopropyl, isobutyl, methoxy, ethoxy, isopropoxy, and isobutoxy; R3c is selected from the group consisting of en, methyl, ethyl, -CH20H, methoxy, ethoxy, and isopropoxy; R3ct and R3e are independently selected from the group consisting of hydrogen, methyl, and ethyl.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', , VII-B', VIII', ', ', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a ceutically acceptable salt thereof, wherein R5 and R8a are ndently selected from the group consisting of hydrogen and methyl.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of ae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', or or a pharmaceutically acceptable salt thereof, n Z is ] In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', or a pharmaceutically acceptable salt thereof, n Z is N-.
In another ment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a ceutically acceptable salt thereof, wherein R7a is selected from the group consisting ofhydrogen and methyl. In another embodiment, R7a is hydrogen. In another embodiment, R7a is methyl.
In another ment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', ', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein: Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S( 0)-, -S( 0)r, , and H(R8a)-; tis I; and R4a and R4b are independently selected from the group consisting of hydrogen and ; or R4a and R4b taken together with the carbon atom to which they are ed form a 3-membered cycloalkyl. In another embodiment, Xis -CHr. In another embodiment, Xis selected from the group consisting of: In another embodiment, Xis . In another embodiment, Xis -S-. In another ment, Xis -CH2S-.
In another embodiment, Xis -N(H)CHr. In another embodiment, Xis selected from the group consisting In r embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R11h is hydrogen.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", , VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R7b is hydrogen.
In r embodiment, sed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', , VIII', VIII-a', ', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R7a and R7b are en.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", , VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, R6b is selected from the group consisting of hydrogen, -Cl, -OMe (or -OCH3), and -OH.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, , IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a ceutically acceptable salt thereof, wherein R9f is hydrogen.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, , IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R9f is methyl.
In r embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically able salt thereof, wherein R11a is selected from the group consisting of hydrogen and -OH.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R11h is hydrogen.
In r embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically able salt thereof, wherein R7b is R7b-l. In r embodiment, R10a and R10b are independently optionally substituted C1_6 alkyl. In another embodiment, R10a and R10b are independently ally substituted Ci-4 alkyl.
] In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', , VIII', ', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", ", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R7b is R7b-2, and PG is BOC. In another embodiment, R10a and R10b are independently optionally substituted C1_6 alkyl. In another embodiment, R10a and R10b are independently optionally substituted C1_4 alkyl.
In another embodiment, disclosed herein is a compound having any one of Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a pharmaceutically acceptable salt thereof, wherein R7b is R7b 3. In another embodiment, m is 2 or 3, and R10a and R10b are each optionally substituted C1_6 alkyl. In another ment, mis 2. In another embodiment, R10a and R10b are independently optionally substituted C1_4 alkyl.
In another embodiment, disclosed herein is a compound having Formulae VIII, or a ceutically acceptable salt thereof, which is any one of the compounds of Table VI.
Table VI .. ,,0 -,,,Q-s ,,,o7 0 0 0 H2N F i= i= i= ····-��oOH �S Y- NH2 0 0 i= F _ 123 - HO z 0-... ,.-- ... .... . .,,0- 11 .,,0 h ·:�7 µ 0 ·:� 0 H2N I NHz �I -<::::,;::; ..,,o ,& Cl ( _av ; and 0 In another ment, disclosed herein is a compound having Formulae VIII, or a pharmaceutically acceptable salt thereof, which is any one ofthe compounds of Table VI-A.
Table VI-A 0 0 E F 0 0 E F 0 0 Lo•··· H,N OyO H2N O C I OH I yCr �:..OH 0 C # 0 0 0 yO... QII• La,,.• "'"Vo � . .
I H2N O 0 OH # V I yCr�: 0 0 0 Lo••·· ''' �:.
H,N O"'° H2N O OH ff . c I V I 0 0 Lo••" o,,.
'' " ' O' H,N O"'° � I H2N Off v OH # V I 0 0 Lo•"· o,,.
' O' H,N OJ)" OH H2N Off # OH 0 0 o,,. o,,.
Lo••·· Lo,..· I O H,N v I O OH "',()'' # F F 0 0 O • • · ,,.L O "' H 2 N � H2N ,;:;:, OH 0 0 O • • · ,•.L o,· ·· H, N H2N I OH 0 0 H 2 N I H 2 N . OH 0 0 H 2 N �2-�:. OH H 2N ,;:;:, OH 0 0 0• • · -Lo•" H 2 N I H 2N � OH OH ,;:;:, 0 0 O • • · o, , .
O" ' " '' L O ' /) ---::: H 2 NO N 1 if H OH HN ON 2 H OH 0 d 0 ---::: HNO.N2 H OH 0 do 0 �:...
---::: HNO.2 N H OH In another embodiment, disclosed herein is a compound having a VII-A or Formula VII-B, or a pharmaceutically acceptable salt thereof, which is any one of the compounds of Table VI-B.
Table VI-B 0 E �,,•OL_ ,,· OJ)'''.LQ\''0 OH H2N ::,,. OH NH2 and In another embodiment, disclosed herein is a compound having Formulae VIII, or a pharmaceutically acceptable salt thereof, ed from the group consisting of: In another embodiment, disclosed herein is a compound having Formula IX, or a pharmaceutically acceptable salt thereof, which is any one ofthe compounds of Table VII.
Table VII i= i= 0 (rS ..,� I I 'Q.;:::, ,,,o NH2 0 i= p OH su OH l ,,(X:S h ..,07· .... o NH2 ,,,o 9' ,,,o � 0 0 i= F OH s OH 0 0 NH2 F i= ..,oT" ,,,o °o 0 NH2 F _,.,N H OH OH _,.,NH _,.,NH ' ' OH OH 0 0 i= i= NH2 H N � 0 .,,0,.,,VA NH2 ' F 0 0 F F i= ; and .... o . � ll �NH2 .,,o ',, In another embodiment, disclosed herein is a compound having Formula IX-a, or a pharmaceutically able salt thereof, selected from the group consisting of: ..07,,, � ,07,, .,,o I ✓,; ...o I 0 � 9" 1 i= 'o � 9" I _,.... NH ; and NH2 V. Methods of making immunoconjugates and synthetic intermediates ] The general synthesis of the immunoconjugates of the disclosure is described in General Scheme 1.
General Scheme 1 ection hydrolysis In General Scheme 1, 7a)(R7b) is a glucocorticosteroid having an -NH(R7a) group (wherein R7a is hydrogen or C1_4 alky), or a nd having any one of Formulae VII, VIII, VIII-a, VIII-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX­ a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", VIII-b", IX", IX-a", or IX-b", or a compound of Table 9; HS-A is an antibody or antigen binding fragment having a limited number of reduced interchain disulfide bonds, R' and R" are independently any side chain found in a naturally-occurring, e.g., methyl, isopropyl, and/or non-natural amino acid, e.g., 2CH2N(H)C(=O)NH2, mis 1, 2, 3, 4, 5, or 6, and PG is a protecting group, e.g., BOC. For the purpose of illustration, General Scheme 1 shows only one sulfhydryl as being available for ation in the antibody or antigen binding fragment.
In another embodiment, disclosed herein is a method of making a compound having a I-c: or a pharmaceutically acceptable salt thereof, wherein: AisA1 orA2; A1 is an anti-tumor necrosis factor (TNF) alpha protein; A2 is a protein; L is a linker; ] n is 1-10; and SM is a radical of a glucocorticosteroid, e.g., a compound having any one of Formulae 11-a-q; the method comprising: a) conjugating a nd having Formula X: SM-L-N� with an anti-tumor necrosis factor (TNF) alpha protein or a protein; and b) isolating the compound having Formula 1-c, or a pharmaceutically acceptable salt thereof.
In another embodiment, the method r comprises hydrolyzing the compound having Formula 1-c to give a compound having Formula 1-d: In another embodiment, disclosed herein is a method of making a compound having Formula 1-e: ( o S)A SM-L-��� n R7a 0 1-e ] or a pharmaceutically able salt thereof, wherein: AisA1 orA2·' A1 is an anti-tumor necrosis factor (TNF) alpha protein; A2 is a n; L is a linker; R7a is selected from the group ting ofhydrogen and Ci-4 alkyl; n is 1-10; mis 1, 2, 3, 4, 5, or 6; and SM is a radical of a glucocorticosteroid, e.g., a compound having any one of Formulae 11-a-e or 1-q; the method comprising: a) conjugating a compound having Formula XI: SM L N��, mN'( R12 0 XI with an anti-tumor necrosis factor (TNF) alpha n or a protein; and b) isolating the nd having Formula I-e, or a pharmaceutically acceptable salt thereof.
In another embodiment, the method further comprises hydrolyzing the compound having Formula I-e to give a compound having Formula I-f: ( o S)A SM L ��� n R7a I-f.
In another embodiment, disclosed herein is a method of making a compound having Formula I-G: n I-G wherein: A is adalimumab; and n is 1-10, the method comprising: ] a) conjugating Cpd. No. 88: Ox .. I v-o�� , _ _ , ... o,, � H _ , H HN ,,, ,,,,:;. � - o HO O H ,.__ 0 NH OH GN_0 0 Cpd. No. 88 with partially-reduced adalimumab; and b) isolating, e.g., by tography, the compound having a 1-G.
In another embodiment, disclosed herein is a method of making a compound having a 1-H: � �\ f-JN--! � 0 ,,_J " V V,,..(o H NH o sJN 0 ) H O n 1-H wherein: A is adalimumab; and n is 1-10, the method sing hydrolyzing the compound having Formula 1-G to give Formula 1-H.
In another embodiment, disclosed herein is a method of making a nd having Formula 1-G or Formula 1-H, wherein n is 1-7. In another embodiment, n is 1-5. In another embodiment, n is 2-4. In another ment, n is 1. In another embodiment, n is 1.5. In another embodiment, n is 2.
In another embodiment, n is 2.5. In another embodiment, n is 3. In another embodiment, n is 3.5.
In another embodiment, n is 4. In another embodiment, n is 4.5. In another embodiment, n is 5.
In another embodiment, disclosed herein is a compound having Formula 1-H: n 1-H wherein: A is adalimumab; and n is 1-10.
In another embodiment, disclosed herein is a compound having Formula I-H, wherein n is 1-7. In another ment, n is 1-5. In another embodiment, n is 2-4. In another ment, n is 1.
In another embodiment, n is 1.5. In another embodiment, n is 2. In another embodiment, n is 2.5.
In another embodiment, n is 3. In another embodiment, n is 3.5. In another embodiment, n is 4.
In another embodiment, n is 4.5. In another embodiment, n is 5. In another embodiment, n is 5.5.
In another embodiment, n is 6. In another embodiment, n is 6.5. In another embodiment, n is 7.
In another embodiment, n is 7.5. In another embodiment, n is 8.
] According to the present disclosure, glucorticoid receptor agonists can be linked to the antibody, antigen-binding fragment thereof, or anti-TNF alpha proteins via any method and at any location that does not prevent the antibody, antigen-binding fragment thereof, or anti-TNF alpha protein from binding antigen (e.g., TNF alpha) or prevent activity of the glucorticoid or agonist. Methods for achieving such a linkage have been discussed, for example, in Panowski et al., mAbs 6: 34-45 (2014), Jain et al., Pharm. Res. 32: 540 (2015), Mack et al., Seminars in Oncology 41: 637-652 (2014), U.S. Published Application No. 2008/0305044, and U.S. Published Application No. 2011/0097322 each h is herein incorporated by reference in its entirety.
The glucorticoid receptor agonists can be linked to the antibodies, antigen-binding fragments f, or anti-TNF alpha proteins via a natural amino acid, e.g., an amino acid that has a side-chain with a nucleophilic group.
For e, the glucorticoid receptor agonist can be linked to a lysine residue. Methods for conjugation via lysine are known. Such methods include a ep process in which a linker is attached to the antibody, antigen-binding fragment thereof, or anti-TNF alpha protein in a first chemical reaction and then the linker is reacted with the orticoid receptor agonist in a second chemical reaction. In another rmethod, a one-step reaction with a preformed linker-glucocorticoid receptor agonist to form the ate containing the glucocorticoid receptor agonist linked to the antibody, antigen-binding fragment therof, or NF alpha protein.
The glucorticoid receptor agonist can also be linked to a cysteine residue. s for conjugation via cysteine are know. IgG 1 antibodies contain four inter-chain disulfide bonds, and conjugation via cysteine can occur after reduction of these bonds creates sulfhydryls available for conjugation.
The glucorticoid or agonists can be linked to the antibody, antigen-binding fragment thereof, or anti-TNF alpha proteins via site-specific conjugation.
One method of site-specific conjugation is ne-based pecific conjugation. An example of this method has been ed by Junutula et al., Nat. Biotechnol 26: 925-935 (2008); see also Junutula et al., J Immunol. Methods 332: 41-52 (2008), each ofwhich is herein incorporated by reference in its entirety. Using this method, antibodies, antigen-binding fragments thereof or anti-TNF alpha proteins can be engineered with additional nes that provide reactive thiol groups to conjugate glucocorticoid receptor agonist. These publications also provide guidance regarding the ion of reactive cysteins that do not interfere with antigen binding.
Another method of site-specific conjugation makes use of selenocysteine. Selenocysteine is similar to cysteine but conatins a more reactive um atom in place of the sulfur atom in cysteine.
Conditions can be used in which selenocysteines are selectively ted. Hofer et al., Biochemistry 48: 12047-12057 (2009), which is herein incorporated by reference in its entirety, has exemplified this technique.
Another method of site-specific ation makes use of unnatural ammo acids, e.g., acetylphenylalanine (pAcPhe) or para-azido phenylalanine (pAF). Wang et al. Proc. Natl. Acad. Sci. 100: 56-61 (2003), Axup et al., Proc. Natl. Acad. Sci. 109:16101-16106 (2012), and Kem et al., JACS 138: 1430-1445 (2016), each of which is herein incorporated by reference in its entirety, have exemplified this technique.
Another method of site-specific conjugation makes use of enzymatic approaches, e.g., via glycotransferases or transglutaminases. Mutant ransferases can be used to attach a chemically active sugar moiety to a glycosylation site on an antibody, antigen-binding fragment thereof, or anti-TNF alpha protein. Human IgG antibodies contain an N-glycosylation site at residue Asn-297 of the Fe fragment. The glycans attached at this residue can be degalactosylated so that a mutant glycotransferase is capable of transferring o. Boeggeman et al., Bioconjug. Chem. 20: 1228-1236 (2009), which is herein incorporated by reference in its entirety, has exemplified this technique. Transglutaminases, e.g., from Streptoverticillium mobaranse, recognize a glutamine tag, e.g., LLQG, that can be engineered into an NF alpha protein. Jeger et al., Angew Chem. Int. Ed. Engl. 49: 9995-9997 (2010), which is herein incorporated by reference in its entirety, has exemplified this technique.
C-terminal attachment via expressed protein on can also be used. For example, intein mediated C-terminal thioester formation can be used for chemoselective ligation with an NF alpha n containing an N-temrinal cysteine peptide. Chiang et al., J Am. Chem. Soc. 136: 3370-3373 (2014), which is herein orated by reference in its entirety, has exemplified this technique.
Also provided herein are synthetic intermediates, e.g., compounds having Formula X and XI, that useful for the preparation of immunoconjugates.
In one embodiment, the tic intermediate sed herein is a compound having any one of Formulae VII, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", VII-A", VII-B", VIII", VIII-a", ", IX", IX-a", or IX-b", or a pharmaceutically able salt thereof, wherein R7b is selected from the group consisting of -L-H, -L-PG, ] In another embodiment, the synthetic intermediate disclosed herein is a compound having Formula VIII, or a pharmaceutically acceptable salt thereof, which is any one or more of the compounds of Table VIII: Table VIII ....0 ,,,,0- s ,,,o7 0 0 HN F h7b ..,,07s ,,,o 0 0 HN F h7b F i= OH OH ...,o_/Y' �S AI ··••0�A1 ,,,o ·••O R7b R7b N, N, yH yH Cl Cl 0 0 i= F HO N,R7b ,,' -r'0 0 0 .,,o hµ F F b F i= ...,07··,,, .,,o � ....o� I ·••O ,& I 0 7 1 7 1 'N :::,._ 'N :::,._ R7 R7b OH07·•'o<-Q HN-R7b 0 0 '-e '" so s� - o �: U � 07 0 1 _ . . ,,\0 .,,0 0 0 R ' " ' .cr-9 HN-R7b R' " P7 � .. ,o O 0 0 0 ; and N-R7b ( � so __U 'V' wherein R7b is ed from the group consisting of -L-H, -L-PG, 0 o j-L�� j-L-N� o , and o .
In another ment, R7b is selected from the group consisting of: 0 � 0 O <""Nn""N�NJ(: I O µ ; and R7"-6 In another embodiment, R7b is R7b_4_ In another embodiment, R7b is R7b_5_ In another embodiment, R7b is RTu -6_ In another embodiment, R7b is any one ofthe structures of Table IX.
Table IX 0 H � 0 O "----/ '¾. )l__ ,,,N')('- I N�w-Z \ ,,l_,�ylN�NJ( O H )-1/ - I O H 0 µ ' 0 ' 0 0 H = 0 '\,)l___, - I '(' N�N_.l( O H )-J 0 07 ('o (O'i HN)l___ 0 �o � 0'1 °) � o �o � lNJ( \ l___,,,o �o o---.) l___,,,o )-J. 0 ' (' o 0 07 (' o 0 / o ( 0 h f) 0 ofo l_, ( ( N--( o o__} l_,o O o .' / o ....,- o o � 0/'-.......,- � 0/'-.......,- �O o '.
/ H o� o� n �o�0,,,.......____, 0,,,.......____, 0,,,.......____,o'-./'- /"'......,,N 0 �N'{ 0 o O '· /Y"-' 0t? o \_�N---( 0 O '· o '· l o ....____, o � o � o/'-.......,-� 0 �� O O '· / o /'-.......,- o /'-.......,-� �0�o/'....../ 0�N1 0)-._f ' In another embodiment, the synthetic intermediate disclosed herein is a compound having Formulae VIII, or a pharmaceutically acceptable salt thereof, which is any one of the compounds ofTable 'o..J /:( Ll..11' y� O� o\-�:� O �( •••LL gs I O o a=<;IZ 0 -( o� I ZI{ ff •I ILL ,,,/ go 0 lLI • 0 0 I lLI • lLI • 0 0 I I I 0 . ·,o 0 . ·,o b-1 ·,o __ . � b-1_ b-1_ � � IZ(0 Q Q Cl) Q �:r o Q 0 J Q ZI �o Q o o> O \.._J Q \_li �lt 0 :. 0 0 -to ZI O o�_.
J >-<::IZ LL •, 0 I )- 0 . ·,o 0 o-1. _ 0 o o_) \__ o� o OJ: ,...., u..,,.
Ll.. 1•• u. 1,, . ·, o 0� _J \ A o II ZI ZI )•"11 o�_,,,, IZ -{ �o " :1f° o ZI 0 . � 0 . � crj. 0 � . � 0 0 I I 0 0 0 0)- 0 I 0 I I 0 0\..-o � Cl)0 0 0)- 0 0 g� I IZt: IZt: Cl)0 _) IZt: \_ o"'o o_) ) \__� )Jo 0 OJ: o_) o 0 Ott ) 0 0 U..1,.
U.. 11, .wq··,o 6 I ZI .. ,,, -< o In another embodiment, the synthetic ediate disclosed herein is a compound having Formulae VIII, or a pharmaceutically acceptable salt f, which is any one of the compounds ofTable Table X-A 0 0 H H ,Jl--,"ll'IJ, fy i o "'°'' f:o ao�,J,Iy i _j[)" o y-y o i yY° H OH � 0 = H O 0 HO, 0 0 HO� 0 0 r(_t, IN iJ_,,.Q))' /( o . H,..r(o = H 0 o,J_,.' -- N, 0 0 ,,. 0 BocHN �N 0 o -= H -- N, 0 0 Q\\\'' 01•· o ..l -- o .L_ ,- r,r') �,-· 0 0 � � ) O o� y!JL BocHN� 0� y�)LNDJ)''' 0 H o = H 0� 0 °r: ' o )-a O O H I I ' y()lN�N0Nn.D oso,H H = H 0 O 0 0 0,,·· •· L_ ,QO ''' O CJ_ )L N�� N 1 N_}lO H H � yJto H o a O=S o''oH BocHN/'\( �� 0 0 -� o=s,,, OOH and In another embodiment, the synthetic intermediate disclosed herein is a compound having a IX, or a pharmace utically acceptable salt thereof, which is any one ofthe chemical structures of Table XI.
Table XI 0 !: F F H 0-SU ""07··•' ;::,._ / I .o R7b ·••O H o �Su ....o ;::,._ / I .o R7a ·••O H 0 0 F F . ... ., , o o� Q HN- R 7 b ....0 7 ··•· , . , o � 0 0 HN- R 7 b ""0 7 •••, , ., ,o I 0 � 'o � � I ,,.- N , R 7b. ' ...,o ., , o I i= � ' o � I ,,.- N , R 7 b, ' ..,,o . . , o I 0 � F � ' o � I HN , R 7 b. ' 0 ..,,v � .. ,,0 7 � R7b ·••O N' H F F 0 OH S ....o� 7b ,11Q ,, H 0 F OH .,,,v Sr{y 0 �N,R7b """-·••0 ' H F F ; and 0 OH S ...,o� 7 �N,R b .,,o H n R7b is selected from the group consisting of -L-H, -L-PG, , ,,( � 1 ,-,> o , and o .
In another embodiment, R7b is selected from the group consisting of: 0 0 H � O �-- � I N 'l(' N�w-Z O )-.!/ ; and In another embodiment, R7b is R7b_4_ In another embodiment, R7b is R7b_5_ In r embodiment, R7b is RTu -6. In another embodiment, R7b any one ofthe chemical structures of Table IX.
In another embodiment, the synthetic intermediate disclosed herein is a compound having Formula IX, or a pharmaceutically able salt thereof, which is any one of the compounds of Table 0 I . ·,,o '' 0 0 6--/ 'a-Io )- 6-p 0 Qz I ZI ... 0 ZI 0Q Q ZI ZI ZI . ,,, 0 o==s .. ,,, ) .. ,,, o==s . IZ IZ IZ -to :1□ZI :1f°-'toZI �1□ZI 7'" � tr),......., (I) )- o o -<o, ••ILL :1< 0 U,_ 11, o=s... ,, I I '<::: 0 ,,-::; 0 . � LL. 1, 0 I 0 I b�'O 0)-0 0 . ·,o I b� pCl) ZI IZ IZ -to _:ro \_ o ZI ZI � o� f: o o ""' . ,,, \.._J o=s. o IZ �o o"o _) OJ� I 0) o=Q-0 ....., 0 0 LL •, 0 . ·,o 0 - o-1._ . ··,o "b.J_ "b-1.
! QZI � � IZ -z _:ro ZI -< o 0 ZI )...., ZI IZ o o � _,,,, -< o OJ,)....IZ OJ�IZ ZI VI. Methods of use and pharmaceutical compositions Provided herein are conjugates having Formulae I-a and 1-b, and glucocorticoid receptor agonists having Formulae VII, VII-A, VII-B, VIII, VIII-a, VIIl-b, IX, IX-a, or IX-b, or any one of Formulae VII', VII-A', VII-B', VIII', VIII-a', VIII-b', IX', IX-a', IX-b', VII", , VII-B", VIII", VIII-a", ", IX", IX-a", or IX-b" (wherein R7b is hydrogen) that can be used in vitro or in vivo. ingly, also provided herein are compositions, e.g., pharmaceutic! compositions for certain in vivo uses, comprising a conjugate or a glucocorticoid receptor agonist described herein having the desired degree of purity in a physiologically acceptable r, excipient or stabilizer gton's Pharmaceutical Sciences (1990) Mack Publishing Co., Easton, PA). Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed.
The compositions (e.g., ceutical compositions) to be used for in vivo administration can be sterile. This is readily lished by filtration through, e.g., sterile tion membranes. The compositions (e.g., pharmaceutical compositions) to be used for in vivo administration can comprise a preservative.
A ceutical ition comprising a glucocorticoid receptor agonist provided herein can be formulated, for example, as a nasal spray, an inhalation aerosol (e.g., for oral inhalation), or a capsule, tablet, or pill (e.g., for oral administration).
The glucocorticoid receptor agonists provided herein (e.g., an anti-TNF ADC) are compounds, wherein the average number of glucocorticosteroids per antibody (DAR) in the composition is about 1 to about 10. In some embodiments, the e number of glucocorticosteroids per antibody (DAR) in the composition is about 2 to about 6. In some embodiments, the average number of glucocorticosteroids per antibody (DAR) in the composition is about 3 to about 4. In some embodiments, the average number of glucocorticosteroids per antibody (DAR) in the composition is about 3 .1. In some embodiments, the average number of glucocorticosteroids per antibody (DAR) in the composition is about 3.2. In some embodiments, the average number of glucocorticosteroids per antibody (DAR) in the composition is about 3 .3. In some embodiments, the average number of glucocorticosteroids per antibody (DAR) in the ition is about 3.4. In some embodiments, the average number of glucocorticosteroids per antibody (DAR) in the composition is about 3 .5. In some embodiments, the average number of glucocorticosteroids per antibody (DAR) in the composition is about 3.6. In some embodiments, the average number ofglucocorticosteroids per dy (DAR) in the ition is about 3.7. In some ments, the average number of glucocorticosteroids per antibody (DAR) in the composition is about 3.8. In some embodiments, the average number of glucocorticosteroids per antibody (DAR) in the composition is about 3.9.
Glucocorticoid receptor agonists and ceutical compositions compnsmg a glucocorticoid or agonist described herein can be useful in ting cytokine e (in vitro or in vivo) and/or for the treatment of autoimmune or inflammatory diseases. Glucocorticoid receptor agonists and pharmaceutical compositions comprising a glucocorticoid receptor agonist described herein can be used for the treatment of asthma (e.g., bronchial asthma), Crohn's disease (e.g., mild to moderate active Crohn's disease involving the ileum and/or the ascending colon and/or the nance of clinical ion of mild to moderate Crohn's disease involving the ileum and/or the ascending colon for up to 3 months), ulcerative colitis (e.g., for the induction of remission in patients with active, mild to moderate ulcerative colitis), allergic rhinitis (e.g. nasal symptoms associated with seasonal allergic rhinitis and/or perennial allergic rhinitis).
For administration to human patients, the total daily dose of glucocorticoid receptor agonists provided herein is typically in the range of 0.001 mg to 5000 mg, or in the range of 0.01 mg to 1000 mg, depending on the mode of administration. For example, oral administration or intravenous, intramuscular, intra-articular, or peri-articular administration can require a total daily dose of from 0.01 mg to 1000 mg, or from 0.1 mg to 100 mg. The total daily dose can be stered in single or divided doses.
A pharmaceutical composition comprising a conjugate provided herein can be formulated, for example, for intravenous administration or infusion.
Conjugates and pharmaceutical compositions comprising conjugates described herein can be useful in lysing a cell expressing surface TNF-alpha (in vitro or in vivo), for the treatment of diseases or disorders characterized by increased pha (e.g., sead TNF-alpha in synovial fluid), and/or for the treatment of an autoimmune or matory disease.
A pharmaceutical composition comprising a glucocortic receptor agonist or a ate described herein is used for the treatment of rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), psoriatic arthritis (PsA), a spondyloarthropathy such as ankylosing spondylitis (AS) or axial spondyloarthritis (axSpA), adult ' disease (CD), pediatric Crohn's disease, ulcerative colitis (UC), plaque sis (Ps), hidradenitis suppurativa (HS), s, Behcets disease, or psoriasis, including plaque psoriasis.
For administration to human patients, the total daily dose of conjugate provided herein is typically in the range of from 0. 01 µg to 100 mg per kg of body weight, and can be given once or more daily, weekly, monthly or yearly.
The disclosure also provides ments (Embs) 1-209 as particular embodiments. The Formulae and Tables referred to these particular embodiments that are not shown in the embodiment are set forth in the description above.
Embodiment (Emb) 1. A compound having Formula I-a: (SM-L-Q)n-A1 I-a or a pharmaceutically acceptable salt or solvate f, wherein: A1 is an anti-tumor necrosis factor (TNF) alpha protein; L is a linker; Q is a heterobifunctional group or trifunctional group; or Q is absent; n is 1-10; and SM is a radical of a glucocorticosteroid.
Emb 2. The nd of Emb 1, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid.
] Emb 3. The compound of Emb 2, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid selected from the group consisting of: H R SM S-� , SM 0-� ' SM N-� and SM N-�,wherem the sulfur, oxygen, or nitrogen atom is attached directly or ctly to the C- or D-ring of the glucocorticosteroid,and R is Ci4 alkyl.
Emb 4. The compound of Emb 3, or a pharmaceutically acceptable salt or solvate thereof, wherein the sulfur, oxygen, or nitrogen atom is attached directly or indirectly to the D-ring of the glucocorticosteroid.
Emb 5. The compound of Emb 2 or Emb 3, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula II-a, wherein: R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen,halo,and hydroxy; R3 is selected from the group consisting of -CH2OH, -CH2SH, SCH2CN, ,and R3a is selected from the group consisting of hydrogen and C1 4 alkyl; R3b is selected from the group ting of C1 4 alkyl and C1 4 alkoxy; R3c is selected from the group consisting of hydrogen, C1 4 alkyl,-CH2OH,and C1 4 alkoxy; R3ct and R3e are independently selected from the group consisting of hydrogen and Ct4 alkyl; R9a is selected from the group consisting of optionally substituted alkyl,optionally tuted lkyl, optionally substituted aryl, and optionally tuted heteroaryl; R9b is selected from the group consisting ofhydrogen and alkyl; or R9a is: Z xyy R6b \�JR6 R6' R6d '· and R9b is hydrogen or methyl; X is selected from the group consisting of -(CR4aR4b)t 5-, , , -S-, -S(=0)-, -S(=0)r, -NR -CH2S-, , -N(H)C(R8a)(R8b)-, -CR4c=CR4ct -, and -C=C-; or Xis absent; tis I or 2; Z is selected from the group consisting of =CH-, =C(0H)-, and =N-; each R4a and R4b are independently selected from the group consisting ofhydrogen and C1 4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting ofhydrogen and C1 4 alkyl; R5 is selected from the group consisting of hydrogen and C1 4 alkyl; R6\ R6b, R6C, R6ct , and R6e are each independently selected from the group consisting of hydrogen, halo, C1 4 alkyl, C1 4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; R8a and R8b are independently selected from the group consisting of hydrogen and C1 4 alkyl; R11 is selected from the group consisting of hydrogen, halo, C1 4 alkyl, y, thiol, amino, alkylthio, and alkoxy; and =-= ents a single or double bond.
] Emb 6. The compound of Emb 5, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-b.
] Emb 7. The compound of any one of Embs 2-4, or a pharmaceutically acceptable salt or e thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-c, wherein: R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group ting of hydrogen, halo, and hydroxy; R9a is selected from the group consisting of optionally tuted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; and R9b is selected from the group consisting of hydrogen and alkyl; or R9a is: R9b is hydrogen; W is selected from the group consisting of and -S-; X is selected from the group consisting of -(CR4aR4b)t 5 8a)(R8b , , -S-, -S(=0)-, -S(=0h-, -NR -, , -CH20-, -N(H)C(R ) , CR4c=CR4ct -, and -C=C-; or X is absent; t is I or 2; Z is selected from the group ting of =CH-, -, and =N-; each R4a and R4b are independently selected from the group consisting of hydrogen and C1 4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3-to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting of hydrogen and Ci-4 alkyl; R5 is selected from the group consisting of hydrogen and C1 4 alkyl; R6\ R6b, R6C, R6ct , and R6e are each independently ed from the group consisting of en, halo, C1 4 alkyl, C1 4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; R8a and R8b are independently selected from the group consisting of hydrogen and C1 4 alkyl; R11 is selected from the group consisting of hydrogen, halo, C1 4 alkyl, C1 4 haloalkyl, cyano, y, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
Emb 8. The compound of Emb 7, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-d.
Emb 9. The compound of any one of Embs 2-4, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent l of a glucocorticosteroid having Formula 11-e, wherein: R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R9c is selected from the group consisting of hydrogen, C1_4 alkyl, and -C(=0)R9\ R9ct is ed from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted lkyl, optionally substituted aryl, and optionally substituted heteroaryl; R9e is selected from the group consisting of hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally tuted heteroaryl; Wis selected from the group consisting of and -S-; and =-= represents a single or double bond.
] Emb 10. The compound of Emb 9, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-f.
Emb 11. The compound of any one of Embs 7-10, or a pharmaceutically acceptable salt or solvate thereof, wherein Wis -S-.
Emb 12. The compound of any one of Embs 7-10, or a pharmaceutically acceptable salt or solvate f, wherein W is .
Emb 13. A compound having Formula 1-b: (SM-L-Q)n-A2 1-b, or a pharmaceutically acceptable salt or solvate f, wherein: A2 is a protein; L is a linker; Q is a bifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a monovalent l of a glucocorticosteroid having any one of: Formula 11-1, Formula 11-m, Formula 11-n, Formula 11- o, Formula 11-por a 11-q, wherein: R1 is selected from the group consisting of en and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group consisting of -CH2OH, -CH2SH, , l, -SCH2F, -SCH2CF3, hydroxy, -OCH2CN, -OCH2Cl, - l�oYnO R3b /�onR3c R3a O ' 0 l�o, /? �-0 R3d and O-R3e ; R3a is selected from the group consisting of hydrogen and C1_4 alkyl; R3b is selected from the group consisting of C1_4 alkyl and C1_4 alkoxy; R3c is selected from the group consisting of hydrogen, C1_4 alkyl, -CH20H, and C1_4 alkoxy; R3ct and R3e are independently selected from hydrogen and C1_4 alkyl; R6\ R6\ R6C, R6ct , and R6e are each ndently selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; X is ed from the group consisting of -(CR4aR4b)t-, , -S-, -S(=0)-, -S(=0)r, -NR\ , -CH20-, -N(H)C(R8a)(R8b)-, -CR4c=CR4ct -, and -C=C-; or Xis absent; Y2 is selected from the group consisting of - 0-, -S-, and )-; or Y2 is absent; tis 1 or 2; Z is selected from the group consisting of =CR11a- and =N-; each R4a and R4b are independently selected from the group consisting of hydrogen and C1_4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting of hydrogen and C1_4 alkyl; R5 is selected from the group consisting of hydrogen and C1_4 alkyl; R7a is selected from the group consisting ofhydrogen and C1_4 alkyl; R8a and R8b are independently selected from the group consisting of hydrogen and C1_4 alkyl; R9f is ed from the group consisting of hydrogen and C1_4 alkyl; R11a and R11h are independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and ; and =-= represents a single or double bond.
Emb 14. The compound of any one ofEmbs 2-4 or 13, or a pharmaceutically able salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-1, wherein: R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group consisting of -CH20H, -CH2SH, , and . R3a is ed from the group consisting ofhydrogen and C1_4 alkyl; R3b is selected from the group consisting of C1_4 alkyl and C1_4 alkoxy; R3c is selected from the group consisting of en, C1_4 alkyl, -CH20H, and C1_4 ; R3ct and R3e are independently selected from hydrogen and C1_4 alkyl; Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=0)-, -S(=0)r, -NR\ -CH2S-, -CH20-, (R8a)(R8b)-, -CR4c=CR4ct -, and -C=C-; or X is absent; t is 1 or 2; Z is selected from the group consisting of =CR11a- and =N-; each R4a and R4b are independently selected from the group consisting of hydrogen and C1_4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting of hydrogen and C1_4 alkyl; Rs is selected from the group consisting of hydrogen and C1_4 alkyl; R6\ R6C, R6ct , and R6e are each independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; Y2 is selected from the group consisting of , -S-, and -N(R7a)-; or Y2 is absent; R7a is ed from the group consisting of hydrogen and C1_4 alkyl; R8a and R8b are independently selected from the group consisting of hydrogen and C1_4 alkyl; R9f is selected from the group consisting of hydrogen and C1_4 alkyl; R11a and R11b are independently ed from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
Emb 15. The compound ofEmb 14, or a ceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-m.
Emb 16. The compound ofEmb 14, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-n.
Emb 17. The compound of any one of Embs 2-4 or 13, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a lent radical of a orticosteroid having Formula 11-o, wherein: R1 is selected from the group consisting of en and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group ting of -CH20H, -CH2SH, '· R3a is ed from the group consisting of hydrogen and C1_4 alkyl; R3b is ed from the group consisting of C1_4 alkyl and C1_4 alkoxy; R3c is selected from the group consisting of hydrogen, C1_4 alkyl, -CH20H, and C1_4 alkoxy; R3ct and R3e are independently selected from hydrogen and C1_4 alkyl; Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=0)-, -S(=0)r, -NR5-, -CH2S-, , -N(H)C(R8a)(R8b)-, -CR4c=CR4ct -, and -C=C-; or Xis absent; tis 1 or 2; Z is selected from the group consisting of=CR11a- and =N-; each R4a and R4b are independently selected from the group consisting of hydrogen and C1_4 alkyl; or R4a and R4b taken together with the carbon atom to which they are ed form a 3- to 6-membered cycloalkyl; R4c and R4ct are ndently ed from the group consisting of hydrogen and C1_4 alkyl; Rs is selected from the group consisting of hydrogen and C1_4 alkyl; R6\ R6\ R6ct , and R6e are each independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; Y2 is selected from the group consisting of , -S-, and - -; or Y2 is absent; R7a is selected from the group consisting of hydrogen and C1 4 alkyl; R8a and R8b are independently ed from the group consisting of hydrogen and C1 4 alkyl; R9f is selected from the group consisting of en and C1 4 alkyl; R11a and R11h are independently selected from the group consisting of hydrogen, halo, C1 4 alkyl, C1 4 haloalkyl, cyano, y, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
Emb 18. The compound of Emb 17, or a ceutically acceptable salt or solvate thereof, n SM is a lent radical of a glucocorticosteroid having Formula 11-p.
Emb 19. The compound of Emb 17, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid having Formula 11-q.
Emb 20. The compound of any one of Embs 5-19, or a pharmaceutically acceptable salt or solvate thereof, wherein =-= represents a double bond.
Emb 21. The compound of any one of Embs 5-20, or a pharmaceutically acceptable salt or solvate thereof, n R1 is selected from the group consisting ogen and fluoro.
] Emb 22. The compound of any one of Embs 5-21, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is selected from the group consisting ofhydrogen and fluoro.
Emb 23. The compound of any one of Embs 5, 6, or 13-22, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from the group consisting of -CH2OH, , -SCH2Cl, - SCH2F, and hydroxy.
Emb 24. The compound of any one of Embs 5, 6, or 13-22, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from the group consisting of: /'-..._....-o : OH 0 ·, /.., .0 0 R3b /_, .0 R3c 'OH ........,,,. y LJ r7, ........,,,. LJ CO2H R3a O O R3a is selected from the group ting of hydrogen and methyl; R3b is selected from the group consisting of methyl, ethyl, isopropyl, isobutyl, methoxy, ethoxy, isopropoxy, and isobutoxy; R3c is selected from the group consisting of hydrogen, methyl, ethyl, -CH2OH, methoxy, ethoxy, and isopropoxy; R3ct and R3e are independently selected from the group consisting of hydrogen, methyl, and ethyl.
Emb 25. The compound of any one of Embs 5-8 or 11-24, or a pharmaceutically acceptable salt or solvate f, wherein R5 and R8a are ndently selected from the group consisting of hydrogen and methyl.
Emb 26. The compound of any one of Embs 5-8, 11--25, or a pharmaceutically acceptable salt or solvate thereof, wherein Z is =CH-.
Emb 27. The compound of any one of Embs 5-8 or 11-25, or a pharmaceutically acceptable salt or solvate thereof, wherein Z is =N-.
Emb 28. The nd of any one of Embs 5-8 or 11-27, or a pharmaceutically able salt or solvate f, wherein R6\ R6ct, and R6e are hydrogen.
Emb 29. The compound of any one of Embs 13-28, or a pharmaceutically acceptable salt or solvate thereof, wherein Y2 is -N (R7a)-.
Emb 30. The compound of Emb 29, or a pharmaceutically acceptable salt or solvate thereof, wherein R7a is selected from the group consisting of hydrogen and methyl.
Emb 31. The compound of Emb 30, or a pharmaceutically acceptable salt or solvate thereof, wherein R7a is hydrogen.
Emb 32. The compound of Emb 30, or a pharmaceutically acceptable salt or solvate thereof, wherein R7a is methyl.
Emb 33. The compound of any one of Embs 5-8 or 13-32, or a pharmaceutically acceptable salt or solvate f, n: Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -, r, -CH2S-, and -N(H)CH(R8a)-; t is l; R4a and R4b are ndently selected from the group consisting of en and methyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3-membered cycloalkyl; and R8a is selected from the group consisting of hydrogen and methyl.
Emb 34. The compound of Emb 33, or a pharmaceutically acceptable salt or solvate thereof, wherein Xis -CHr.
] Emb 35. The compound of Emb 33, or a pharmaceutically acceptable salt or solvate thereof, wherein X is selected from the group consisting of: Emb 36. The compound of Emb 33, or a pharmaceutically acceptable salt or solvate thereof, wherein X is .
Emb 37. The compound of Emb 33, or a pharmaceutically acceptable salt or solvate f, wherein X is -S-.
Emb 38. The compound of Emb 33, or a pharmaceutically acceptable salt or e thereof, wherein Xis -CH2S-.
Emb 39. The compound of Emb 33, or a pharmaceutically acceptable salt or e thereof, wherein Xis -N(H)CHr.
Emb 40. The compound of Emb 33, or a pharmaceutically acceptable salt or solvate thereof, wherein X is selected from the group consisting of: Emb 41. The compound of any one of Embs 13-16 or 20-40, or a pharmaceutically able salt or solvate thereof, wherein R6c is ed from the group consisting of hydrogen, -Cl, - OCH3, and hydroxy.
Emb 42. The compound of any one of Embs 13 or 17-40, or a pharmaceutically acceptable salt or solvate thereof, wherein R6b is selected from the group consisting of hydrogen, -Cl, -OCH3, and hydroxy.
Emb 43. The compound of any one of Embs 13-42, or a pharmaceutically acceptable salt or solvate thereof, wherein R9f is hydrogen.
Emb 44. The compound of any one of Embs 13-42, or a pharmaceutically acceptable salt or solvate f, n R9f is methyl.
Emb 45. The compound of any one of Embs 13-44, or a pharmaceutically acceptable salt or solvate thereof, wherein R11a is selected from the group ting of hydrogen and hydroxy.
Emb 46. The compound of any one of Embs 13-44, or a pharmaceutically acceptable salt or solvate thereof, wherein R11b is hydrogen.
] Emb 47. The compound of any one of Embs 1-46, or a pharmaceutically acceptable salt or solvate thereof, wherein L is a cleavable .
Emb 48. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate f, wherein the cleavable linker comprises a succinimide, amide, thiourea, thioether, oxime, or self-immolative group, or a combination thereof.
Emb 49. The compound of any one of Embs 1-48, or a pharmaceutically able salt or solvate thereof, n the cleavable linker comprises a peptide.
Emb 50. The compound of Emb 49, or a pharmaceutically acceptable salt or solvate thereof, wherein the cleavable linker comprises a tripeptide.
Emb 51. The compound of Emb 49, or a pharmaceutically acceptable salt or solvate thereof, n the cleavable linker comprises a dipeptide.
Emb 52. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein the ble linker ses phosphate ester.
Emb 53. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein the cleavable linker ses a pyrophosphate diester.
Emb 54. The compound of any one of Embs 1-53, or a pharmaceutically acceptable salt or solvate thereof, wherein Q is absent.
Emb 55. The compound of any one of Embs 1-53, or a pharmaceutically acceptable salt or solvate thereof, wherein Q is a heterobifunctional group selected from the group consisting of Q-1, Q-2, Q-3, Q-4, Q-5, and Q-6, n mis 1, 2, 3, 4, 5, or 6.
Emb 56. The compound of any one of Embs 1-53, or a pharmaceutically acceptable salt or solvate thereof, wherein Q is a heterotrifunctional group that is Q-7.
Emb 57. The compound ofEmb 55, or a ceutically acceptable salt or solvate thereof, wherein Q is selected from the group consisting ofQ-1, Q-2, Q-3, and Q-4.
Emb 58. The compound ofEmb 57, or a pharmaceutically acceptable salt or solvate f, wherein Q is selected from the group consisting of Q-3 and Q-4.
Emb 59. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-1; mis 1 or 2; and R10a and R10b are independently selected from the group ting ofhydrogen and optionally substituted C1_6 alkyl.
Emb 60. The compound ofEmb 59, or a pharmaceutically acceptable salt or solvate thereof, n -L-Q-is LQ-2.
Emb 61. The compound ofEmb 59, or a pharmaceutically able salt or e thereof, wherein -L-Q-is LQ-3.
Emb 62. The compound ofEmb 59, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-4.
Emb 63. The compound ofEmb 59, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-5.
Emb 64. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-6; mis 1 or 2; and R10a and R10b are ndently selected from the group consisting ofhydrogen and optionally substituted C1_6 alkyl.
Emb 65. The compound ofEmb 64, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-7.
] Emb 66. The nd ofEmb 64, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-8.
Emb 67. The compound ofEmb 64, or a pharmaceutically able salt or solvate thereof, wherein -L-Q-is LQ-9.
Emb 68. The compound ofEmb 64, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q- is: LQ-10.
Emb 69. The nd of any one of Embs 1-4 7, or a pharmaceutically acceptable salt or solvate thereof, wherein L is a noncleavable linker.
Emb 70. The compound of any one of Embs 1-47, or a ceutically acceptable salt or solvate thereof, wherein the linker comprises one or more polyethylene glycol units.
Emb 71. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q- is LQ-11; mis 1 or 2; and xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15.
] Emb 72. The nd of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q- is LQ-12; mis 1 or 2; and xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15.
Emb 73. The nd of any one of Embs 1-47, or a pharmaceutically able salt or solvate f, wherein -L-Q- is LQ-14; mis 1 or 2; xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or ; and R10a and R10b are independently selected from the group consisting of en and optionally substituted C1 6 alkyl.
Emb 74. The compound ofEmb 73, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-15.
Emb 75. The compound ofEmb 73, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-16.
Emb 76. The compound ofEmb 73, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-1 7.
Emb 77. The compound ofEmb 73, or a pharmaceutically able salt or solvate thereof, wherein -L-Q-is LQ-18.
Emb 78. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, n -L-Q- is LQ-19; mis 1 or 2; xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or ; and R10a and R10b are independently selected from the group consisting of hydrogen and optionally substituted C1 6 alkyl.
Emb 79. The compound ofEmb 78, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-20.
Emb 80. The nd ofEmb 78, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-21.
Emb 81. The compound ofEmb 78, or a pharmaceutically acceptable salt or solvate thereof, wherein s LQ-22.
Emb 82. The compound ofEmb 78, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-23.
Emb 83. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q- is LQ-13; and xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15.
Emb 84. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q- is LQ-29; and xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15.
Emb 85. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-24; xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; and R10a and R10b are independently selected from the group consisting of hydrogen and optionally substituted C1_6 alkyl.
Emb 86. The compound ofEmb 85, or a pharmaceutically acceptable salt or solvate f, wherein -L-Q-is LQ-25.
Emb 86A. The nd of Emb 85, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q- is LQ-26.
Emb 87. The compound ofEmb 85, or a pharmaceutically acceptable salt or solvate thereof, n -L-Q-is LQ-27.
Emb 88. The nd ofEmb 85, or a ceutically able salt or solvate thereof, wherein -L-Q-is LQ-28.
Emb 89. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q- is LQ-30; xis 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; and R10a and R10b are independently selected from the group ting of hydrogen and optionally substituted C1_6 alkyl.
Emb 90. The compound ofEmb 89, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-31.
Emb 91. The compound ofEmb 89, or a pharmaceutically acceptable salt or solvate f, wherein -L-Q-is LQ-32.
Emb 92. The compound ofEmb 89, or a ceutically acceptable salt or e thereof, wherein -L-Q-is LQ-33.
Emb 93. The compound ofEmb 89, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q-is LQ-34.
Emb 94. The compound of any one of Embs 55, 59-68, or 71-82, or a ceutically acceptable salt or solvate thereof, wherein m is 2.
Emb 95. The compound of any one of Embs 1-47, or a pharmaceutically acceptable salt or solvate thereof, wherein -L-Q- is any one the chemical structures of Table I.
Emb 96. The compound of any one of Embs 1-95, or a pharmaceutically acceptable salt or solvate thereof, wherein n is 2-8.
Emb 97. The compound of Emb 96, or a pharmaceutically acceptable salt or solvate thereof, wherein n is 2-5.
Emb 98. The compound of any one of Embs 1-95, or a pharmaceutically acceptable salt or solvate thereof, wherein n is 2.
Emb 99. The compound of any one of Embs 1-95, or a ceutically acceptable salt or solvate thereof, n n is 4.
Emb 100. The compound of any one of Embs 1 or 47-99, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid which is any one of the chemical structures of Table II.
] Emb 101. The compound of Emb 100, or a pharmaceutically acceptable salt or solvate thereof, wherein SM is a monovalent radical of a glucocorticosteroid selected from the group consisting ..,,07-,,,, ·••O I o ·:�;,,•,,� a i" r- I 'o 'o ::::,.._ N HN,/ _,, .._/ and Emb 102. The compound of any one of Embs 1-101, or a pharmaceutically able salt or solvate f, wherein A1 is an antibody or antigen-binding fragment thereof or wherein A2 is an antibody or antigen-binding fragment f.
Emb 103. The compound of any one of Embs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is an anti-tumor is factor (TNF) alpha protein that binds to human TNF alpha and/or murine TNF alpha or wherein A2 is protein that binds to human TNF alpha and/or murine TNF alpha.
Emb 104. The compound of any one of Embs 1-101, or a ceutically acceptable salt or solvate thereof, wherein A1 is an anti-tumor necrosis factor (TNF) alpha protein that binds to soluble TNF alpha or n A2 is a protein that binds to soluble TNF alpha.
Emb 105. The compound of any one of Embs 1-101, or a pharmaceutically acceptable salt or e thereof, wherein A1 is an anti-tumor necrosis factor (TNF) alpha protein that binds to membranebound TNF alpha or wherein A2 is a protein that binds to ne-bound TNF alpha.
Emb 106. The compound of any one of Embs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is an anti-tumor necrosis factor (TNF) alpha protein comprising an anti-TNF antibody or wherein A2 is protein comprising an anti-TNF antibody.
Emb 107. The nd of any one of Embs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is an anti-tumor is factor (TNF) alpha protein comprising an antigenbinding fragment of an anti-TNF antibody or wherein A2 is a protein comprising an antigen-binding fragment of an anti-TNF antibody.
Emb 108. The compound of any one of Embs 102-105 or 107, or a pharmaceutically able salt or solvate thereof, wherein the antigen-binding fragment is selected from the group consisting of Fab, Fab', F(ab')2, single chain Fv or scFv, disulfide linked Fv, V-NAR , lgNar, intrabody, lgGACH2, minibody, F(ab')3, ody, triabody, diabody, single-domain antibody, DVD-lg, Fcab, mAb2, (scFv)2, or scFv-Fc.
] Emb 109. The compound of any one of Embs 1-108, or a pharmaceutically acceptable salt or solvate thereof,wherein the antibody or antigen-binding fragment thereof is murine, chimeric, zed, or human.
Emb 110. The compound of any one of Embs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A 1 is an umor necrosis factor (TNF) alpha protein comprising a soluble TNF receptor or wherein A2 is a protein comprising a soluble TNF receptor.
Emb 111. The compound of Emb 110, or a pharmaceutically acceptable salt or solvate thereof, wherein the e TNF receptor is a soluble p75 TNF receptor.
Emb 112. The compound of any one of Embs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 comprises a heavy chain constant domain or a fragment thereof or wherein or A2 comprises a heavy chain constant domain or a fragment thereof.
Emb 113. The compound of Emb 112, or a ceutically acceptable salt or solvate thereof, wherein the heavy chain constant domain or fragment thereof comprises a constant domain ed from the group consisting of: (a) an lgA constant domain; (b) an lgD constant domain; (c) an lgE constant domain; (d) an lgGl constant domain;(e) an lgG2 constant ; (f) an lgG3 constant domain; (g) an lgG4 constant domain; and (h) an lgM constant domain or is a fragment thereof.
Emb 114. The compound of Emb 113, or a pharmaceutically acceptable salt or solvate thereof, wherein the heavy chain nt domain comprises a human lgG1 heavy chain constant domain or fragment thereof.
Emb 115. The compound of Emb 114, or a pharmaceutically acceptable salt or solvate thereof, wherein the heavy chain constant domain comprises a human lgG 1 Fe domain.
Emb 116. The nd of any one of Embs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 comprises a light chain constant domain or a nt thereof or n A2 comprises a light chain constant domain or a fragment thereof.
] Emb 117. The compound of Emb 116, or a pharmaceutically acceptable salt or solvate thereof, wherein the light chain constant domain or fragment thereof ses a constant domain selected group consisting of (a) an lg kappa constant domain and (b) an lg lambda constant domain or is a fragment thereof.
Emb 118. The compound of any one of Embs 1-101, or a ceutically acceptable salt or solvate thereof, wherein A1 competitively inhibits binding of an antibody selected from the group ting of adalimumab, imab, certolizumab pegol, and golimumab to TNF-alpha or wherein A2 competitively inhibits binding of an antibody selected from the group consisting of adalimumab, infliximab, izumab pegol, and golimumab to TNF-alpha.
Emb 119. The compound ofany one ofEmbs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 binds to the same TNF-alpha epitope as an antibody selected from the group consisting of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab or wherein A2 binds to the same TNF-alpha epitope as an antibody selected from the group consisting of adalimumab, imab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab.
Emb 120. The compound ofany one ofEmbs 1-101, or a pharmaceutically acceptable salt or solvate thereof, n the anti-TNF alpha n or protein is selected from the group consisting of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab.
] Emb 121. The compound ofany one ofEmbs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 comprises the variable heavy chain CDR l, CDR2, and CDR3 ces of SEQ ID NO:3 or 6, SEQ ID NO:4, and SEQ ID NO:5, tively and the variable light chain CDRl, CDR2, and CDR3 sequences ofSEQ ID NO:32, SEQ ID NO:33, and SEQ ID NO:34, respectively or wherein A2 comprises the variable heavy chain CDRl, CDR2, and CDR3 sequences ofSEQ ID NO:3 or 6, SEQ ID NO:4, and SEQ ID NO:5 respectively and the variable light chain CDRl, CDR2, and CDR3 sequences ofSEQ ID NO:32, SEQ ID NO:33, and SEQ ID NO:34, respectively.
Emb 122. The compound ofany one ofEmbs 1-101, or a pharmaceutically acceptable salt or e thereof, wherein A1 comprises the variable heavy chain sequence of SEQ ID NO:50 and the variable light chain sequence of SEQ ID NO:59 or n A2 comprises the variable heavy chain sequence ofSEQ ID NO:50 and the variable light chain sequence ofSEQ ID NO:59.
Emb 123. The compound ofany one ofEmbs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1does not bind to TNF beta or wherein A2 does not bind to TNF beta.
Emb 124. The compound ofany one ofEmbs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 binds to TNF beta or wherein A2 binds to TNF beta.
Emb 125. The compound ofany one ofEmbs 1-101, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 neutralizes human TNF-alpha cytotoxicity in a standard in vitro L929 assay with an IC50 of 1x10 7 Mor less or wherein A2 neutralizes human TNF-alpha cytotoxicity in a standard in vitro L929 assay with an IC50 of1x10 7 Mor less.
Emb 126. The compound ofany one ofEmbs 1-101, or a ceutically able salt or solvate thereof, wherein A1 blocks the interaction ofTNF-alpha with p55 and p75 cell surface receptors or wherein A2 blocks the ction ofTNF-alpha with p55 and p75 cell surface receptors.
Emb 127. The compound of any one of Embs 1-101, or a pharmaceutically acceptable salt or e thereof, wherein A1 lyses surface TNF expressing cells in vitro in the presence of complement or wherein A2 lyses surface TNF expressing cells in vitro in the presence of complement.
Emb 128. The compound of Emb 111, or a pharmaceutically acceptable salt or solvate thereof, wherein the soluble p75 TNF receptor is cept.
Emb 129. The compound of Emb 102, or a ceutically acceptable salt or solvate thereof, wherein the dy is adalimumab.
Emb 130. The compound of any one of Embs 1-101, wherein A1 binds to Fe gamma receptor or wherein A2 binds to Fe gamma receptor.
Emb 131. The compound of any one of Embs 1-101, n A1 is active in the GRE embrane TNF-alpha reporter assay and/or the L929 assay or wherein A2 is active in the GRE transmembrane TNF-alpha reporter assay and/or the L929 assay.
Emb 132. The compound ofany one ofEmbs 1 or 102-131, or a ceutically acceptable salt or solvate thereof, which is any one of the chemical structures of Table III, wherein n is 1-5 and A is A1 or A2.
Emb 133. The compound of Emb 132, or a pharmaceutically acceptable salt or solvate thereof, which is any one of the chemical structures ofTable IV, wherein A is A1 or A2.
Emb 134. The compound ofany one ofEmbs 1 or 102-131, or a pharmaceutically acceptable salt or solvate thereof, which is any one of the chemical structures of Table V, wherein n is 1-5 and A is A1 or A2.
Emb 135. The compound of Emb 134, or a pharmaceutically acceptable salt or e thereof, wherein n is 2 or 4.
Emb 136. A pharmaceutical composition comprising the compound of any one of Embs 1-135, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically able carrier.
Emb 13 7. A pharmaceutical composition comprising a plurality of the nds of any one ofEmbs 1-135 or a pharmaceutically salt or solvate thereof, wherein the compounds in the pharmaceutical composition have an average of 1 to 10 SM-L-Q per A1 or A2, i.e., n is 1-10, 2 to 6 SM-L-Q per A1 or A2, 3 to 4 SM-L-Q per A1 or A2, about 2 SM-L-Q per A1 or A2, about 3 SM-L-Q per A1 or A2, or about 4 SML-Q per A1 or A2.
Emb 138. A method for lysing a cell sing e TNF-alpha comprising contacting the cell with the compound ofany one ofEmbs 1-135 or the pharmaceutical composition ofEmbs 136 or 137.
Emb 139. A method for treating an autoimmune disease in a patient in need thereof comprising administering to said patient the compound of any one of Embs 1-135 or the pharmaceutical composition ofEmbs 136 or 137.
Emb 140. The method ofEmb 139, wherein said autoimmune disease is rheumatoid arthritis, juvenile thic arthritis, psoriatic arthritis, ankylosing spondylitis, adult Crohn's disease, pediatric Crohn's disease, ulcerative colitis, plaque psoriasis, enitis suppurativa, uveitis, Behcets disease, a spondyloarthropathy, or psoriasis.
Emb 141. A method for treating a e or disorder characterized by increased TNF-alpha in al fluid in a patient in need f comprising administering to said patient the compound ofany one of Embs 1-135 or the pharmaceutical composition ofEmbs 136 or 137.
Emb 142. A compound having Formula VII, or a pharmaceutically acceptable salt or solvate thereof, wherein: R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of en, halo, and hydroxy; R3 is selected from the group ting of -CH20H, - CH2SH, -CH2Cl, -SCH2Cl, , -SCH2CF3, hydroxy, -OCH2CN, -OCH2Cl, -OCH2F, -OCH3, - /�o�OH 0 l�oYno y·,,oH R3b C02H R3a 0 and /�o, /? �-o-R3d O-R3e ; R3a is selected from the group consisting of hydrogen and C1_4 alkyl; R3b is selected from the group consisting of C1_4 alkyl and C1_4 alkoxy; R3c is ed from the group consisting of hydrogen, C1_4 alkyl, -CH2OH, and C1_4 alkoxy; R3ct and R3e are independently selected from hydrogen and C1_4 alkyl; X is selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=O)-, -S(=O)r, -NR5-, - CH2S-, -CH2O-, -N(H)C(R8a)(R8b)-, -CR4c=CR4ct -, and -C=C-; or Xis absent; tis 1 or 2; Z is selected from the group consisting of =CR11a- and =N-; each R4a and R4b are independently selected from the group consisting of hydrogen and C1_4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting ogen and C1_4 alkyl; R5 is selected from the group ting ofhydrogen and C1_4 alkyl; R6\ R6b, R6C, and R6ct are each independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, v haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; R7a is selected from the group consisting ofhydrogen and C1_4 alkyl; R7b is selected from the group consisting of hydrogen, -L-H, -L-PG, 0 0 j-L-N� j-L¾Y 0 , and O ; mis 1, 2, 3, 4, 5, or 6; Lis a linker; PG is a protecting group; R9f is selected from the group ting of hydrogen and C1_4 alkyl; R8a and R8b are independently selected from the group consisting of hydrogen and C1_4 alkyl; R11a and R11b are independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, Cn haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= ents a single or double bond.
] Emb 143. The compound of Emb 142, or a pharmaceutically acceptable salt or solvate thereof, wherein R7b is selected from the group ting of R7b-l, R7b-2, and R7b-3; mis 1, 2, 3, 4, 5, or 6; and R10a and R10b are each independently selected from the group consisting of hydrogen and optionally substituted C1 6 alkyl.
Emb 144. The compound of Embs 142 or 143, or a pharmaceutically acceptable salt or solvate thereof, having Formula VIII.
Emb 145. The compound of Emb 144, or a pharmaceutically acceptable salt or e thereof, having a VIII-a.
Emb 146. The compound of Emb 144, or a pharmaceutically acceptable salt or solvate thereof, having Formula VIII-b.
Emb 147. The compound ofEmb 142 or 143, or a pharmaceutically acceptable salt or solvate thereof, having Formula IX.
Emb 148. The compound of Emb 147, or a pharmaceutically acceptable salt or solvate thereof, having a IX-a.
Emb 149. The compound of Emb 147, or a pharmaceutically acceptable salt or solvate thereof, having Formula IX-b.
Emb 150. The compound of any one of Embs 142-149, or a pharmaceutically acceptable salt or solvate thereof, n =-= ents a double bond.
Emb 151. The compound of any one of Embs 142-150, or a ceutically acceptable salt or solvate thereof, wherein R1 is selected from the group consisting of hydrogen and fluoro.
] Emb 152. The compound of any one of Embs 142-151, or a pharmaceutically able salt or solvate thereof, wherein R2 is selected from the group consisting of hydrogen and fluoro.
Emb 153. The compound of any one of Embs 142-152, or a pharmaceutically able salt or solvate thereof, wherein R3 is selected from the group consisting of -CH20H, -CH2Cl, -SCH2Cl, - SCH2F, and hydroxy.
Emb 154. The compound of any one of Embs 142-152, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is ed from the group consisting of: R3a is selected from the group consisting of hydrogen and methyl; R3b is selected from the group consisting of methyl, ethyl, isopropyl, isobutyl, methoxy, ethoxy, isopropoxy, and isobutoxy; R3c is selected from the group consisting of hydrogen, , ethyl, -CH20H, methoxy, ethoxy, and isopropoxy; R3ct and R3e are independently selected from the group consisting of hydrogen, methyl, and ethyl.
] Emb 155. The compound of any one of Embs 142-154, or a pharmaceutically acceptable salt or solvate thereof, wherein R5 and R8a are independently selected from the group consisting of hydrogen and methyl.
Emb 156. The compound of any one of Embs 142-155, or a pharmaceutically acceptable salt or solvate thereof, wherein Z is =CH-.
Emb 157. The compound of any one of Embs 142-155, or a pharmaceutically able salt or solvate thereof, n Z is =N-.
Emb 158. The compound of any one of Embs 142-155, or a pharmaceutically acceptable salt or e thereof, wherein R7a is selected from the group consisting of hydrogen and methyl.
] Emb 159. The compound of Emb 158, or a ceutically acceptable salt or e thereof, wherein R7a is hydrogen.
Emb 160. The compound of Emb 158, or a ceutically acceptable salt or solvate thereof, wherein R7a is methyl.
Emb 161. The compound of any one of Embs 142-160, or a pharmaceutically acceptable salt or solvate f, n: Xis ed from the group consisting of -(CR4aR4b)t-, , -S-, -S(=O)-, -S(=O)r, -CH2S-, and -N(H)CH(R8a)-; t is l; and R4a and R4b are independently selected from the group consisting of hydrogen and methyl; or R4a and R4b taken er with the carbon atom to which they are attached form a 3-membered cycloalkyl.
Emb 162. The compound of Emb 161, or a pharmaceutically acceptable salt or solvate thereof, wherein Xis -CH2-.
Emb 163. The compound of Emb 161, or a pharmaceutically acceptable salt or solvate thereof, wherein X is selected from the group consisting of: Emb 164. The compound of Emb 161, or a pharmaceutically acceptable salt or solvate thereof, wherein Xis .
Emb 165. The compound of Emb 161, or a pharmaceutically acceptable salt or solvate thereof, wherein Xis -S-.
Emb 166. The compound of Emb 161, or a pharmaceutically acceptable salt or solvate thereof, wherein Xis -CH2S-.
] Emb 167. The compound of Emb 161, or a pharmaceutically acceptable salt or solvate thereof, wherein Xis -N(H)CHr.
] Emb 168. The compound of Emb 161, or a pharmaceutically acceptable salt or solvate thereof, wherein X is selected from the group consisting of: Emb 169. The compound of any one of Embs 142-168, or a pharmaceutically acceptable salt or solvate thereof, wherein R11h is hydrogen.
Emb 170. The compound of any one of Embs 9, or a pharmaceutically acceptable salt or e thereof, wherein R7b is hydrogen.
Emb 171. The compound of any one of Embs 142-170, or a pharmaceutically acceptable salt or solvate thereof, wherein R6b is selected from the group consisting of hydrogen, -Cl, -OCH3, and hydroxy.
Emb 172. The compound of any one of Embs 142-171, or a pharmaceutically acceptable salt or solvate thereof, wherein R9f is hydrogen.
Emb 173. The compound of any one of Embs 142-171, or a pharmaceutically acceptable salt or solvate thereof, wherein R9f is .
Emb 174. The compound of any one of Embs 142-173, or a pharmaceutically acceptable salt or solvate f, wherein R11a is selected from the group consisting of hydrogen and hydroxy.
Emb 175. The nd of any one of Embs 4, or a ceutically acceptable salt or solvate thereof, wherein R11h is hydrogen.
Emb 176. The compound of any one of Embs 143-175, or a pharmaceutically acceptable salt or e thereof, wherein R7b is R7b-1.
] Emb 177. The compound of Emb 176, or a pharmaceutically acceptable salt or solvate thereof, wherein R10a and R10b are independently optionally substituted C1_6 alkyl.
Emb 178. The compound of any one of Embs 143-175, or a pharmaceutically acceptable salt or solvate thereof, wherein R7b is R7b-2, and PG is BOC.
Emb 179. The compound of Emb 178, or a pharmaceutically acceptable salt or solvate thereof, wherein R10a and R10b are independently optionally substituted C1_6 alkyl.
Emb 180. The compound of any one of Embs 143-175, or a ceutically acceptable salt or solvate thereof, wherein R7b is R7b-3.
Emb 181. The compound of Emb 180, or a ceutically acceptable salt or e thereof, wherein mis 1 or 2, and R10a and R10b are each optionally substituted C1_6 alkyl.
Emb 182. The compound of Emb 142, or a pharmaceutically acceptable salt or solvate thereof, which is any one or more of the compounds ofTable VI.
] Emb 183. The nd of Emb 182, or a pharmaceutically acceptable salt or solvate thereof, selected from the group ting of: ( oQ""' "o�]' , and F Emb 184. The compound of Emb 142, or a pharmaceutically acceptable salt or solvate thereof, which is any one or more of the chemical structures of Table VIII, wherein R7b is selected from the group consisting ofRTu -4, R7b-5, andRTu -6.
Emb 185. The compound of Emb 184, or a pharmaceutically acceptable salt or solvate thereof, wherein R7b is R7b-4.
Emb 186. The compound of Emb 184, or a pharmaceutically acceptable salt or solvate thereof, whereinR7b is R7b_5_ Emb 187. The compound of Emb 184, or a pharmaceutically acceptable salt or solvate thereof, wherein R7b is R7b-6.
Emb 188. The compound of Emb 142, or a pharmaceutically acceptable salt or solvate thereof, which is any one or more ofthe chemical structures of Table VIII, whereinR7b is any one ofthe chemical structures of Table IX.
Emb 189. The compound of Emb 142, or a pharmaceutically acceptable salt or solvate f, which is any one of the compounds of Table X.
Emb 190. The compound of Emb 142, or a pharmaceutically able salt or solvate thereof, which is any one of the compounds of Table VII.
Emb 191. The compound of Emb 190, or a pharmaceutically acceptable salt or solvate thereof, ed from the group consisting of: OH OH H2Ny') �,)---o ��0 OH ,,.. NH and Emb 192. The compound of Emb 142, or a ceutically acceptable salt or solvate thereof, which is any one of the nds of Table XI, wherein R7b is selected from the group consisting ofR7b-4, R7b-5 and R7b-6.
Emb 193. The compound of Emb 192, or a pharmaceutically acceptable salt or solvate f, wherein R7b is R7b-4.
Emb 194. The compound of Emb 192, or a pharmaceutically acceptable salt or solvate thereof, n R7b is R7b_5_ Emb 195. The compound of Emb 192, or a pharmaceutically acceptable salt or e thereof, wherein R7b is R7b-6.
Emb 196. The compound of Emb 142, or a pharmaceutically acceptable salt or solvate f, which is any one of the chemical structures of Table XI, wherein R7b any one of the structures of Table IX.
] Emb 197. The compound of Emb 142, or a pharmaceutically acceptable salt or solvate thereof, which is any one of the compounds of Table XII.
Emb 198. A pharmaceutical composition comprising the compound of any one ofEmbs 142- 197, or a pharmaceutically acceptable salt or solvate thereof, wherein R7b is hydrogen, and a pharmaceutically acceptable carrier.
Emb 199. A method for treating an autoimmune or inflammatory disease in a patient in need thereof, the method comprising administering to said patient the compound of any one ofEmbs 142- 197, or a pharmaceutically acceptable salt or solvate thereof, wherein R7b is hydrogen, or the pharmaceutical composition ofEmb 198.
Emb 200. The method of Emb 199, wherein said autoimmune disease is rheumatoid arthritis, juvenile idiopathic arthritis, psoriatic arthritis, ankylosing spondylitis, adult Crohn's disease, pediatric Crohn's disease, ulcerative colitis, plaque sis, or hidradenitis ativa.
] Emb 201. A method of making a compound having Formula 1-c, or a pharmaceutically acceptable salt or solvate thereof, n: A1 is an anti-tumor necrosis factor (TNF) alpha protein; L is a linker; n is 1-1 0; and SM is a radical of a glucocorticosteroid, the method comprising: a) ating a compound having Formula X with an anti-tumor necrosis factor (TNF) alpha protein; and b) isolating the compound having Formula 1-c, or a pharmaceutically able salt or solvate thereof.
Emb 202. The method ofEmb 201 further comprising hydrolyzing the compound having Formula le to give a compound having Formula 1-d.
Emb 203. A method of making a compound having Formula 1-e, or a pharmaceutically acceptable salt or solvate thereof, wherein: A1 is an anti-tumor necrosis factor (TNF) alpha protein; L is a linker; R7a is selected from the group consisting of hydrogen and C1_4 alkyl; n is 1-10; mis 1, 2, 3, 4, 5, or 6; and SM is a radical of a glucocorticosteroid, the method comprising: a) ating a compound having Formula XI, with an anti-tumor necrosis factor (TNF) alpha protein; and b) isolating the compound having Formula 1-e, or a ceutically able salt or solvate thereof.
Emb 204. The method ofEmb 203 further comprising hydrolyzing the compound having Formula 1-e to give a compound having Formula 1-f.
] Emb 205. The compound ofEmb 182, which is 0 � .,,, 1.•'� y ,,Q NH2 or a pharmaceutically acceptable salt or solvate thereof.
Emb 206. The compound ofEmb 182, which is o s ,,,107·' u� .,,o or a pharmaceutically able salt or solvate thereof.
Emb 207. The compound ofEmb 182, which is ( 6--Q p7 .. , NH2 or a pharmaceutically acceptable salt or solvate thereof.
Emb 208. The compound ofEmb 189, which is \ �N-t � 0 � '" V V,,,.(o H \ �I 0 vo HO or a pharmaceutically acceptable salt or solvate thereof.
Emb 209. The compound of Emb 132, or a pharmaceutically acceptable salt or solvate thereof, which is any one of the chemical structures e IV,wherein A is A1 or A2.
The disclosure also provides Embs 1-XXXIII as particular embodiments. The Formulae and Tables referred to these particular embodiments that are not shown in Embs 1-XXXII are set forth in the description above.
Emb I. A compound having Formula I-a: Q)n-A1 I-a wherein: A1 is an anti-tumor necrosis factor (TNF) alpha protein; L is a linker; Q is a heterobifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a monovalent l of a glucocorticosteroid.
] Emb II. A compound having Formula 1-b: (SM-L-Q)n-A2 1-b wherein A2 is a protein; Lis a linker; Q is a heterobifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a l of a glucocorticosteroid having Formula 11-m or Formula 11-p; R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and y; R3 is selected from the group consisting of -CH2OH, -CH2SH, -CH2Cl, - SCH2Cl, -SCH2F, -SCH2CF3, hydroxy, -OCH2CN, l, -OCH2F, -OCH3, H3, -SCH2CN, /�o OH= 0 ., I, ,0 'OH ...._.,,,- y n O R 3b I, ,0 R 3c /�o... /? y, ...._.,,,- n �-o-R3d C02H R38 0 0 , and O -R3e ; R 3a is selected from the group consisting of hydrogen and C1_4 alkyl; R3b is selected from the group consisting of C1_4 alkyl and C1_4 alkoxy; R3c is selected from the group consisting of hydrogen, C1_4 alkyl, , and C1_4 alkoxy; R3ct and R3e are independently selected from hydrogen and C1_4 alkyl; R6\ R6b, R6C, R6ct , and R6e are each independently ed from the group consisting ofhydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and ; Xis ed from the group consisting of -(CR4aR4b)t-, , - S-, -S( O)-, -S( O)r, -NR5-, -CH2S-, -CH2O-, -N(H)C(R8a)(R8b)-, -CR4c CR4ct -, and -C C-; or X is absent; Y2 is selected from the group ting of , -S-, and -N(R7a)-; or Y2 is absent; tis 1 or 2; Z is selected from the group consisting of CRIta- and N-; each R4a and R4b are independently selected from the group consisting of hydrogen and C1_4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3-to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting of hydrogen and C1_4 alkyl; R5 is selected from the group consisting of hydrogen and C1_4 alkyl; R7a is selected from the group consisting of hydrogen and C1_4 alkyl; R8a and R8b are independently selected from the group consisting of hydrogen and C1_4 alkyl; R9f is selected from the group consisting of hydrogen and C1_4 alkyl; R11a and R11h are independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
Emb III. The compound of Embs I or II, wherein SM is a radical of a orticosteroid having Formula 11-m; R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group ting of -CH20H, -CH2SH, , -SCH2Cl, -SCH2F, -SCH2CF3, y, -0CH2CN, -0CH2Cl, -0CH2F, -0CH3, - /�o�OH y·,,0H l�oYYo R3b 0CH2CH3, -SCH2CN, C02H R3a 0 and l�o-- /? �-o-R3d O-R3e ; R3a is selected from the group consisting of hydrogen and C1_4 alkyl; R3b is selected from the group consisting of C1_4 alkyl and C1_4 alkoxy; R3c is selected from the group ting of hydrogen, C1_4 alkyl, -CH20H, and C1_4 alkoxy; R3ct and R3e are independently selected from en and C1_4 alkyl; R6\ R6C, R6ct , and R6e are each ndently selected from the group consisting of hydrogen, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, y, thiol, amino, alkylthio, and alkoxy; Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=0)-, -S(=0)r, -NR5-, -CH2S-, -CH20-, -N(H)C(R8a)(R8b)-, -CR4c=CR4ct -, and -C=C-; or Xis absent; Y2 is ed from the group ting of , -S-, and -N(R7a)­ ; or Y2 is absent; t is I or 2; Z is =CH-; each R4a and R4b are independently selected from the group consisting of hydrogen and C1_4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting ofhydrogen and C1_4 alkyl; R5 is selected from the group consisting ofhydrogen and C1_4 alkyl; R7a is selected from the group consisting of hydrogen and C 1_4 alkyl; R8a and R8b are independently ed from the group consisting ofhydrogen and C1_4 alkyl; R9f is selected from the group consisting of hydrogen and C1_4 alkyl; R11b is selected from the group consisting of en, halo, C1_4 alkyl, C1_4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
Emb IV. The compound of Embs II or III, wherein =-= represents a double bond; R1 is selected from the group consisting of hydrogen and fluoro; R2 is selected from the group consisting of hydrogen and fluoro; R3 is selected from the group consisting of -CH20H, -CH2Cl, -SCH2Cl, -SCH2F, and l�o, l? �-o-R3d 0-R36 R3ct and R3e are independently selected from the group consisting of hydrogen, methyl, and ethyl; R6\ R6C, R6ct , and R6e are hydrogen;Xis selected from the group consisting of -CHr, - 0-, -S-, -S(=0)-, -S(=0)r, -CH2S-, and -N(H)CHr;Y2 is -N(H)-; Z is =CH-; R9f is en; and R11b is hydrogen.
Emb V. The compound of any one of Embs I-IV, wherein L is a linker comprising a dipeptide.
Emb VI. The compound of any one of Embs 1-V, wherein Q is a heterobifunctional group selected from the group consisting of Q-3 and Q-4 and m is 1, 2, 3, or 4.
Emb VII. The compound of any one of Embs I-VII, wherein -L-Q- is LQ-7; mis 2 or 3; and R10a and R10b are independently selected from the group consisting of en and C1 4 alkyl.
Emb VIII. The compound of any one of Embs I-VII, wherein n is 2-5.
Emb IX. The compound of Embs I or II, wherein SM is a monovalent l of a orticosteroid which is any one of the compounds of Table II.
Emb X. The compound of any one of Embs I or III-IX, wherein A1 is (i) an antibody or antigen-binding fragment thereof that binds to human TNF alpha or (ii) a e TNF receptor.
Emb XI. The compound of any one of Embs I or III-X, wherein A1 is ed from the group consisting of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab.
Emb XII. The compound of Emb I, which is any one or more of the compounds of Table III, wherein n is 1-5; A is A1; and A1 is selected from the group consisting of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab.
Emb XIII. The compound of Emb II, which is any one or more of the compounds of Table III, wherein n is 1-5; A is A2; and A2 is selected from the group consisting of antibody, an antigen-binding fragment thereof, or a e receptor protein.
Emb XIV. A compound ed from the group consisting of: \ �N-( � 0 � � '=- V o H 0 NH 0 A sJN� \_H HO O �- O H O e/ _,___)l A S � OH --c �\N'I� �-01•·--· 0 0 0 H O ,, , _,___)l A s--{� �\N'I� OH wherein n is 1-5 and A is an antibody comprising the heavy and light chain sequences of SEQ ID N0:66 and SEQ ID N0:73, respectively.
Emb XV. The compound of Emb XIV selected from the group consisting of: Structure n A 4 A 2 .l ,-· ,,. 2 O O O 0 A S--c��� � � OH ] Emb XVI. The compound ofEmb XIV, wherein the compound is Structure n Emb XVII. The compound ofEmb XIV, wherein the compound is Structure n Emb XVIII. The compound ofEmb XIV, wherein the compound is Structure n Emb XIX. The nd ofEmb XIV, wherein the compound is Structure n Emb. XX. The compound ofEmb XIV, wherein the compound is Structure n ,,,.l 4 O O O 0 A S--c� � -r � OH Emb. XXI. The compound ofEmb XIV, wherein the compound is Structure n .l ,-· ,,. 2 O O O 0 A S--c� � \ -r � OH Emb XXII. A pharmaceutical composition comprising the compound of any one of Embs 1-XXI, and a ceutically acceptable carrier.
Emb XXIII. A method for treating an autoimmune disease in a patient in need f comprising administering to said t the compound of any one of Embs 1-XXI or the pharmaceutical composition of Emb XXII, optionally wherein said autoimmune disease is rheumatoid arthritis, juvenile idiopathic arthritis, tic arthritis, ankylosing spondylitis, adult Crohn's disease, ric s disease, ulcerative colitis, plaque psoriasis, hidradenitis suppurativa, uveitis, Behcets disease, a spondyloarthropathy, or psoriasis.
Emb XXIV. A compound having Formula VII, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group consisting of - CH20H, -CH2SH, -CH2Cl, -SCH2Cl, , -SCH2CF3, -CH20S(=0)20H, hydroxy, -OCH2CN, /�o�OH y·,,oH l�o Y'r(o R3b -OCH2Cl, -OCH2F, -OCH3, H3, -SCH2CN, C02H R3a 0 I ....... c /�o.... ,9 "' o'r(R3 �-0 R3d 0 , and O-R3e ; R3a is ed from the group consisting of hydrogen and Ci-4 alkyl; R3b is selected from the group consisting ofC1_4 alkyl and C1_4 alkoxy; R3c is selected from the group consisting of hydrogen, C1_4 alkyl, -CH20H, C1_4 alkoxy, mino), and -CH2CH2C(=O)OR3f; R3ct and R3e are independently selected from the group consisting of en and C1_4 alkyl; R3f is selected from the group consisting of hydrogen and C1_4 alkyl; Xis selected from the group consisting of -(CR4aR4b)t-, - 0-, -S-, -S(=O)-, -S(=O)r, -NR5-, -CH2S-, -CH20-, -N(H)C(R8a)(R8b)-, -CR4c=CR4ct -, -C=C-, -N(R5)C(=O)-, and -OC(=O)-; or X is ; t is I or 2; Z is selected from the group consisting of =CR11a- and =N-; each R4a and R4b are independently selected from the group ting of hydrogen and C1_4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting of hydrogen and C1_4 alkyl; R5 is selected from the group consisting of hydrogen and C1_4 alkyl; R6\ R6b, R6C, and R6ct are each independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, haloalkyl, cyano, y, thiol, amino, alkylthio, and alkoxy; R7a is selected from the group consisting of hydrogen and �-L¾,Ny C1_4 alkyl; R7b is selected from the group consisting of hydrogen, -L-H, -L-PG, 0 , and j-L-N> 0 or R7a and R7b taken together with the nitrogen atom to which they are attached form: 0 ; or R7a and R7b taken together with the nitrogen atom to which they are attached form a nitro (-NO2) group; mis 1, 2, 3, 4, 5, or 6; Lis a ; PG is a protecting group; R9f is selected from the group consisting of hydrogen and C1_4 alkyl; R8a and R8b are independently selected from the group consisting of hydrogen and C1_4 alkyl; R11a and R11b are independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, Cn haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond Emb XXV. A compound having Formula VII-A or Formula VII-B: 0 R3 rzrx�6bl7b -yy _�N R7a ·•10 11b R60 . R VII-A or R2 VII-B, or a pharmaceutically able salt or solvate thereof, wherein: ] R1 is selected from the group ting of hydrogen and halo; R2 is ed from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group consisting of , -CH2SH, -CH2Cl, -SCH2Cl, -SCH2F, - SCH2CF3, -CH2OS(=O)iOH, hydroxy, -OCH2CN, -OCH2Cl, -OCH2F, -OCH3, -OCH2CH3, -SCH2CN, /�o�OH H /�oYo R3b /�o n R3c C02H R3a O O R3a is selected from the group consisting ofhydrogen and Ci-4 alkyl; R3b is selected from the group consisting of C1_4 alkyl and Ci-4 alkoxy; R3c is selected from the group consisting of en, C1_4 alkyl, -CH2OH, C1_4 alkoxy, -CH2(amino), and -CH2CH2C(=O)OR3f; R3ct and R3e are independently selected from the group consisting of en and Ci-4 alkyl; R3f is selected from the group consisting of hydrogen and C1_4 alkyl;X is selected from the group consisting of-(CR4aR4b)c, , -S-, -S(=O)-, -S(=O)r, -NR\ -CH2S-, -CH2O-, -N(H)C(R8a)(R8b)-, -CR4c=CR4ct -, -C=C-, -N(R5)C(=O)-, and -OC(=O)-; or X is absent; tis 1 or 2; Z is ed from the group consisting of CR11a- and N-; each R4a and R4b are independently selected from the group consisting of hydrogen and Ci-4 alkyl; or R4a and R4b taken together with the carbon atom to which they are ed form a 3- to 6-membered cycloalkyl; R4c and R4ct are independently selected from the group consisting of hydrogen and C1 4 alkyl; R5 is selected from the group consisting ofhydrogen and C1_4 alkyl; R6\ R6b, and R6c are each independently selected from the group consisting of hydrogen, halo, C1_4 alkyl, haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; R7a is selected from the group consisting ofhydrogen and Ci-4 alkyl; R7b is selected from the group consisting of hydrogen, -L-H, -L-PG, 0 , and j-L-N� 0 ; or R7a and R7b taken together with the nitrogen atom to which they are attached form: ] mis 1, 2, 3, 4, 5, or 6; L is a linker; PG is a protecting group; R9f is selected from the group consisting ofhydrogen and Ci-4 alkyl; R8a and R8b are independently selected from the group consisting ofhydrogen and Ci-4 alkyl; ] R11a and R11b are independently selected from the group ting of hydrogen, halo, C1_4 alkyl, Cn haloalkyl, cyano, y, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
Emb XXVI. The compound ofEmbs XXIV or XXV, or a pharmaceutically acceptable salt or solvate thereof, wherein R7b is selected from the group ting of R7b - 1, R7b-2, and R7b-3; mis 1, 2, 3, 4, 5, or 6; and R10a and R10b are each independently selected from the group consisting of hydrogen and optionally substituted C1-6 alkyl.
EmbXXVII. The compound of Embs XXIV or XXVI, or a pharmaceutically able salt or e thereof, having Formula VIII-a.
Emb XXVIII. The compound ofany one ofEmbs XXIV-XXVII, or a pharmaceutically acceptable salt or solvate thereof, wherein =-= represents a double bond; R1 is selected from the group consisting of hydrogen and fluoro; R2 is selected from the group consisting ofhydrogen and fluoro; R3 is I'--._,,,,a, /? �-o-R3d selected from the group consisting of -CH20H, -CH2Cl, -SCH2Cl, -SCH2F, and O-R39 ; R3ct and R3e are independently selected from the group ting of hydrogen, methyl, and ethyl; Z is CH-; R6\R6b, R6C, andR6ct are hydrogen; R7a is hydrogen;Xis selected from the group consisting , , -S-, -S( O)-, -S( O)r, -CH2S-, and Hr;R9f is hydrogen; andR11b is en.
Emb XXIX. The compound of any one of Embs XXIV-XXVIII, or a pharmaceutically acceptable salt or solvate thereof, whereinR7b is hydrogen.
Emb XXX. The compound of any one of Embs XXIV-XXVIII, or a pharmaceutically acceptable salt or solvate thereof, whereinR7b is R7b-1.
Emb XXXI. The compound of any one of Embs XXIV-XXVIII, or a pharmaceutically acceptable salt or solvate thereof, whereinR7b is RTu -2, and PG is BOC.
EmbXXXII. The compound ofany one ofEmbs XXIV-XXVIII, or a ceutically acceptable salt or e thereof, whereinR7b is R7b-3.
Emb XXXIII. The compound of Emb XXIX, or a ceutically acceptable salt or solvate thereof, which is any one or more ofthe compounds ofTableVI.
EmbXXXIV. The compound of Emb XXIX, or a pharmaceutically acceptable salt or solvate thereof, which is any one ofthe compounds ofTableVII.
] V. The compound ofEmb XXXIII, or a pharmaceutically acceptable salt or solvate thereof, which is: EmbXXXVI. The compound of Emb XXIV, or a pharmaceutically acceptable salt or solvate f, which is any one or more of the nds ofTableVIII , wherein R7b is selected from the group consisting ofRTu -4, R7b-5, andRTu -6.
EmbXXXVII. The compound of EmbXXIV, or a pharmaceutically acceptable salt or solvate thereof, which is any one or more ofthe compounds ofTableX.
EmbXXXVIII. The nd ofEmb XXXVII, or a pharmaceutically acceptable salt or solvate thereof, which is: \ r!N-( � 0� � � V V,,,.(o H _r-/ NH o 0 { )----r- 0 �o HO o EmbXXXIX. A method ofmaking a compound having Formula 1-e: O S A SMLN��, mN'{ n R7a 0 or a pharmaceutically acceptable salt or solvate thereof, wherein A is A1 or A2; A1 is an anti-tumor necrosis factor (TNF) alpha protein; A2 is a protein;L is a linker; R7a is selected from the group consisting of hydrogen and C1 4 alkyl; n is 1-10; mis 1, 2, 3, 4, 5, or 6; and SM is a l of a glucocorticosteroid, the method comprising: a) ating a compound having Formula XI: SM L N�p, mN'{ R12 0 XI with an anti-tumor necrosis factor (TNF) alpha protein or a protein; and b) isolating the compound having Formula I-e, or a pharmaceutically able salt or solvate f.
EmbXL. The method of Emb XXXIV further compnsmg hydrolyzing the compound having Formula I-e to give a compound having Formula I-f: 0 s A SM-L-�-¼, R?a 2 n or a ceutically acceptable salt or solvate thereof.
EmbXLI. A compound which is: wherein A is adalimumab.
Emb XLII. A ition comprising the compound ofEmb XLI.
] Embodiments of the present disclosure can be further defined by reference to the following non-limiting examples, which describe in detail preparation of certain antibodies of the present disclosure and methods for using antibodies of the present sure. It will be apparent to those skilled in the art that many modifications, both to materials and methods, can be practiced without ing from the scope of the present disclosure.
Examples It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this disclosure.
Analytical Methods for Compound sis and Characterization Analytical data is included within the procedures below, in the illustrations of the general procedures, or in the tables of examples. Unless otherwise stated, all 1H and 13C NMR data were collected on a Varian Mercury Plus 400 MHz or a Bruker AVIII 300 MHz instrument; chemical shifts are quoted in parts per n (ppm). HPLC analytical data are either detailed within the mental or referenced to the table of LC/MS and HPLC conditions, using the method provided in Table 7.
Table 7: List of LC/MS and GC/MS Methods Method Conditions The gradient was 10-100% Bin 3.4 min with a hold at 100% B for 0.45 min, 100-10% B in 0.0lmin, and then held at 10% B for 0.65 min (0.8 mL/min flow rate). Mobile phase A was 0.0375% trifluoroactic acid in water, mobile phase Bwas 0.018% TFA in MeCN.
The column used for the chromatography was a 2.0 x 50 mm phenomenex Luna-Cl8 column (5 µm particles). Detection methods are diode array (DAD) and ative light scattering (ELSD) detection as well as positive electrospray ionization(MS).
The gradient was 1-90% Bin 3.4 min, 90-100% Bin 0.45 min, 100-1% Bin 0.01 min, and then held at 1 % B for 0.65 min (0.8 mL/min flow rate). Mobile phase A was 0.0375% CF 3CO2H in water, mobile phase Bwas 0.018% CF3CO2H in CH3CN. The column used for the chromatography was a 2.0 x 50 mm enex Luna-Cl8 column (5 µm particles). Detection methods are diode array (DAD) and evaporative light scattering (ELSD) detection as well as positive electrospray ionization (MS).
The gradient was 10-100%Bin 3.4 min with a hold at 100% B for 0.45 min, 100-10%B in 0.0lmin, and then held at 10% B for 0.65 min (0.8 mL/min flow rate). Mobile phase A was 0.0375% CF 3CO2 H in water, mobile phase Bwas 0.018% CF 3CO2H in CH3CN. The column used for the chromatography was a 2.0 x 50 mm phenomenex l8 column (5 µm particles). Detection methods are diode array (DAD) and evaporative light scattering (ELSD) detection as well as positive electrospray ionization (MS).
The gradient was 5% B for0.2 min, and to 95%Bwithin 1.7 min then with a hold at 95% B for 1.3 min,back to 5% B within 0.01min (2.3 mL/min flow rate). Mobile phase A was 0.01% TFA in water, and mobile phase Bwas 0.01 % TFA in HPLC grade MeCN.
The column used for the chromatography was an XBridge Cl8 column (4.6 x 50 mm, 3.5µm particles). Detection methods are diode array (DAD) and ative light scattering (ELSD) ion as well as positive/negative electrospray ionization The gradient was 5% Bto 95%B within 1.5 minthen with a hold at 95%B for 1.5 min, back to 5% Bwithin 0.01 min (2.3 mL/min flow rate).
Mobile phase A was 10 mM NH4HCO3 in water, and mobile phase Bwas HPLCgrade MeCN. The column used for the chromatography was a XBridge Cl8 column (4.6 x 50 mm, 3.5µm particles). Detection methods are diode array (DAD) and evaporative light ring (ELSD) ion as well as ve/negative electrospray ionization.
Mobile Phase: A: Water (0.01 % TFA); B: MeCN (0.01% TFA). Gradient: 5% Bincrease to 95%B within1.2 min, 95%B for 1.3 min,back to 5% B 0.01 min, at a flow rate of2.0 mL/min. Column: SunFire Cl8 (4.6 x 50 mm, 3.5 □m). Column Temp: 50 °C Detection: UV (214, 254 nm) and MS (ESI, Pos mode, 110 to 1000 amu) The gradient was 5% B for0.1 min, and to 95%Bwithin 1.0 min then with a hold at 95% g B for0.9 min, back to 5% B within0.01 min (3.0 mL/min flow rate). Mobile phase A was 0.05% TFA in water, and mobile phase Bwas 0.05% TFA in HPLCgrade MeCN. The column used for the chromatography was a Zorbax SB-C 18 Rapid Resolution HT column (4.6 x 30 mm,1.8 µm particles). Detection methods are diode array (DAD) and evaporative light scattering (ELSD) detection as well as positive/negative electrospray ionization.
Mobile Phase: A: water (0.1% TFA); B: MeCN (0.1 % TFA). Gradient: 5% B increase to h 95% B within 1.3 min,95% B for 1.5 min,back to 5% B within 0.01 min, at a flow rate of 2 . Column: Sunfire Cl8 (4.6 x 50 mm,3.5 µm). Column Temperature: 50 °C Mobile Phase: A: water (0.01% TFA); B: MeCN (0.01% TFA). Gradient: 5% B for 0.2 crease to 95% B within 1.5 min,95% B for 1.5 ck to 5% B within 0.01 min,at a flow rate of 2 mL/min. Column: Sunfire (50 x 4.6 mm,3.5 µm). Column Temperature: 50 °C Mobile phase: A: water (0.05% TFA); B: MeCn (0.05% TFA). Gradient: 5% increase to 100% ofB in 1.3 min, at a flow rate of 2 mL/min. Column: SunFire Cl8 (4.6 x 50 mm, 3.5 µm). Detection: UV (214, 254 nm) and MS (ESI,Pos mode,110 to 1000 amu).
Column Temperature: 50 °C Mobile Phase: A: water (10 mM NH4HCO3); B: MeCN. Gradient: 5% increase to 95% B k in 1.5 min, at a flow rate of 1.8 mL/min. Column: XBridge Cl8 (4.6 x 50 mm,3.5 µm).
Column Temperature: 50 °C Mobile phase: A: water (10 mM NH4HCO3); B: MeCN. Gradient: 10% increase to 95% ofB in 1.5 min,at a flow rate of 1.8 mL/min. Column: Xbridge Cl8(2) (4.6 x 50 mm,3.5 µm). Column Temperature: 50 °C Detection: UV (214,254 nm) and MS (ESI,Pos mode,103 to 800 amu) Mobile Phase: A: Water (0.01%TFA) B: MeCN (0.01% TFA). 5% B increase to 95% B within 1.2 min,95% B for 1.3 min,back to 5% B within n. Flow Rate: 2.0 . Column: e Cl8,4.6*50mm,3.5□m. Column Temperature: 50 °C.
Detection: UV (214, 4 nm) and MS (ESI,Pos mode,110 to 1000 amu).
The gradient was 10-100% B in 3 .4 min with a hold at 100% B for 0.45 0-10% B in 0.01 min,and then held at 10% B for 0.65 min (0.8 mL/min flow rate). Mobile phase A was 0.0375% TFA in water. Mobile phase B was 0.018% TFA in CH3CN. The column used for the chromatography was a Phenomenex Luna-Cl8 column (2.0 x 50 mm, µm particles). Detection methods are diode array (DAD) and evaporative light scattering (ELSD) ion as well as positive electrospray ionization (MS).
A nt of5-100% MeCN (A) and 10 mM ammonium acetate in water (B) was used, 0 at a flow rate of1.5 mL/min (0-0.05 min 5% A,0.05-1.2 min 5-100% A,1.2-1.4 min 100% A,1.4-1.5 min 100-5% A. 0.25 min post-run delay). 2-coupled C8 5 um IOOA Waters e columns (30mm x 75mm each). A gradient of MeCN (A) and 10 mM ammonium acetate in water (B) was used, at a flow rate of q 50mL/min (0-0.5 min 5% A, 0.5-8.5 min linear gradient X to Y% A, .7 min 100% A, 10.7-11 min linear gradient 100-05% A). Linear gradient is stated in the synthetic procedure ofthe compound.
A gradient of5-100% MeCN (A) and 0.1 % TFA in water (B) was used, at a flow rate of r 1.5 mL/min (0-0.05 min 5% A, 0.05-1.2 min 5-100% A, 1.2-1.4 min 100% A, 1.4-1.5 min 100-5% A. 0.25 min un delay).
Analytical UPLC-MS was performed on a Waters SQD mass spectrometer and y UPLC system running MassLynx 4.1 and Openlynx 4.1 software. The SQD mass spectrometer was operated under positive APCI ionization ions. s The column used was a Waters BEH C8, 1.7µm (2.1mm x 30mm) at a temperature of 55°C. A nt of 10-100% acetonitrile (A) and 10 mM ammonium acetate in water (B) was used, at a flow rate of l.OmL/min (0-0.1 min % A, 0.1-1.1 min 10-100% A, 1.1-1.3 min 100% A, 1.3-1.4 min 100-10% A).
Abbreviations used in the examples that follow are: APCI Atmospheric pressure chemical HIC Hydrophobic Interaction tography Bn Benzyl HPLC High performance liquid chromatography BOC tert-butyloxycarbonyl IBX 2-Iodoxybenzoic acid BSA Bovine serum albumin MeCN Acetontrile Cbz Carbobenzyloxy MeOH Methanol CuCN Copper cyanide MgSO4 Magnesium sulfate D2O Deuterated water Min Minute(s) DAD Diode array MP-NaCNBH3 Sodium cynaoborohydride on solid support DCM Dichloromethane MTBE Dimethyl methyl tert-butyl ether DIAD Diisopropyl azodicarboxylate NaCN Sodium cyanide DIPEA N, N-Diisopropylethylamine NaHCO3 Sodium hydrogen carbonate DMA Dimethylacetamide NaHSO3 Sodium hydrogen sulfate DMF Dimethyl formamide Na2SO4 Sodium sulfate DMSO Dimethyl ide NMR r magnetic resonance EiC Extracted ion chromatogram Pd2dba3 tris(dibenzylideneacetone)dipalladium(O) ELSD Evaporative light scattering PBST Phosphate Buffered Saline with Tween 20 detector Eq Equivalent PE Petroleum ether Et2O Diethyl ether PPh3 Triphenyl ine EtOAc Ethyl acetate FMOC 9-Fluorenylmethyloxycarbonyl RP Reverse phase H Hour(s) Rt Retention time H2SO4 Sulfuric acid TBAF Tetrabutylammonium flouride HATU 1- TBS-Cl tert-Butylchlorodimethylsilane [Bis( dimethylamino lene]- IH-1,2,3-triazolo[4,5- b ]pyridinium 3-oxid hexafluorophosphate HCl Hydrochloric acid TFA Trifluoroacetic acid HEPES 4-(2-hydroxyethyl)-l- TLC Thin layer chromatography piperazineethanesulfonic acid Example 1: Synthesis of (2S,6aS,6bR,7S,8aS,8bS,I OR, l laR,12aS, 12bS)-l0-( 4-(4- henoxy)pheny1) difluorohydroxy-8b-(2-hydroxyacetyl )-6a, 8a-dimethyll ,2,6a, 6b,7,8,8a,8b, Ila,12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone Step 1: Synthesis of tert-butyl ( 4-(4-formylphenoxy)phenyl)carbamate ffo,b Boe 000 I h' F 'N CHO To a solution of tert-butyl (4-hydroxyphenyl)carbamate (10 g, 47.8 mmol) and 4-fluorobenzaldehyde (11.86 g, 96 mmol) in N,N-dimethylformamide (100 mL) was added potassium carbonate (39.6 g, 287 mmol). The mixture was stirred at 90°C for 5 hours. One additional vial was set up as described above. All two reaction mixtures were combined and diluted with DCM (300 mL), then ted with water (3 X 100 mL). The organic layer was washed with brine (100 mL) and dried over Na2S04, filtered and concentrated under reduced pressure. The residue was ed by column chromatography (eluted with PE: EtOAc = 30:1 to 5:1) to obtain the target compound (20 g, 63.8 mmol, 66.7 % yield) as a yellow solid. 1H NMR (400MHz, DMSO-d 6) 8 9.91 (s, lH), 9.45 (s, lH), 7.90 (d, J=8.6 Hz, 2H), 7.54 (d, J=8.8 Hz, 2H), 7.11 -7.02 (m, 4H), 1.48 (s, 9H).
Step 2: Synthesis of (2S,6aS,6bR 7S,8aS,8bS, 1 OR, l laR,12aS,12bS)- l 0-( 4-( 4- aminophenoxy)pheny1) -2, 6b-difluorohydroxy-8b-(2 -hydroxyacetyl)-6a, 8a-dimethyll ,2, 6a,6b,7,8,8a,8b, l 12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[ l,2-d][l,3]dioxolone E E 0 0 ('Yo� Boe, � � HO••· N CHO HO••· A suspension of (6S,8S,9R,10S,11S,13S,14S,16R,17S)-6,9-difluoro-l l,16,17-trihydroxy-l 7- (2-hydroxyacetyl)-l imethyl-6,7,8,9,10,11, 12, 13,14, 15,16, l7-dodecahydro-3H- cyclopenta[a]phenanthrenone (4.5 g, 10.91 mmol) and magnesium sulfate (6.57 g, 54.6 mmol) in MeCN (100 mL) was allowed to stir at 20°C for 1 hours A on of tert-butyl (4-(4- formylphenoxy)phenyl)carbamate (3.42 g, 10.91 mmol) in MeCN (100 mL) was added in one portion.
Trifluoromethanesulfonic acid (4.84 mL, 54.6 mmol) was added se via syringe while maintaining an internal temperature of 25°C using an ice bath. After the addition, the mixture was stirred at 20°C for 2 hours. Three onal vials were set up as described above. All four reaction mixtures were combined and filtered, the filtrate was concentrated under reduced pressure to give a residue, which was purified by Prep-HPLC to obtain the target compound (7.5 g, 12.34 mmol, 28.8 % yield) as a yellow solid. LCMS (Method a, Table 7) R1 = 2.21 min; MS m/z = 608.3 (M+H)\ 1H NMR (400MHz, DMSO-d 6) 8 7.36 (d, J=8.6 Hz, 2H), 7.27 (d, J=l0.lHz, lH), 6.85 (d, J=8.6 Hz, 2H), 6.75 (d, J=8.6 Hz, 2H), 6.58 (d, J=8.6 Hz, 2H), 6.29 (dd, J=l.3, 10.1 Hz, lH), 6.13 (s, lH), 5.76 - 5.65 (m, lH), 5.62 - 5.57 (m, lH), 5.54 (d, J=3.l Hz, lH), 5.44 (s, lH), 5.12 (t, J=5.8 Hz, lH), 5.00 (s, 2H), 4.94 (d, J=4.9 Hz, lH), 4.53 (dd, J=6.4, 19.4 Hz, lH), 4.26 - 4.14 (m, 2H), 2.72 - 2.58 (m, lH), 2.34 - 2.17 (m, 2H), 2.04 (d, J=l3.7 Hz, lH), 1.77 - 1.62 (m, 3H), 1.49 (s, 3H), 0.86 (s, 3H). Prep-HPLC Method: Instrument: Gilson 281 semi-preparative HPLC system, Mobile phase: A: CF3CO2H/H2O = 0.075% v/v; B: CH3OH; Column: Phenomenex Luna Cl8250*50mm*10 um; Flow rate: 80 mL/min; Monitor wavelength: 220 & 254 nm.
Time 0.0 20.0 20.1 20.2 30.2 30.3 31.5 B% 28 58 58 100 100 28 28 Example 2: Synthesis of (2S,6aS,6bR,7S,8aS,8bS,1OS, l laR,12aS,12bS)- l 0-(4-(3-aminobenzyl)phenyl)- ifluorohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- ,6b,7,8,8a,8b, l la,12,12a, 12bdodecahydro-4H-naphtho [2', l ':4,5]indeno[1,2-d][1,3]dioxolone Step 1: Synthesis of 4-(bromomethyl)benzaldehyde NC OHC � � �Br - �Br Diisobutylaluminum hydride (153 mL, 153 mmol, 1 Min toluene) was added drop-wise to a 0 �C solution of 4-(bromomethyl)benzonitrile (20 g, 102 mmol) in toluene (400 mL over 1 hour Two additional vials were set up as described above. All three reaction mixtures were combined. The mixture on was added 10% s HCl (1.5 L). The mixture was extracted with DCM (3 X 500 mL).
The organic layer was dried over Na2SO4, filtered and concentrated under reduced re. The residue was purified by column chromatography on silica gel ( eluted with Ac = 10/1) to obtain the target compound (50 g, yield 82%) as white solid. 1H NMR (400MHz, chloroform-d) 8 10.02 (s, lH), 7.91 - 7.82 (m, 2H), 7.56 (d, J=7.9 Hz, 2H), 4.55 - 4.45 (m, 2H).
] Step 2: Synthesis of 3-(4,4,5,5-Tetramethyl- l,3,2-dioxaborolanyl)aniline To a solution of 3-bromoaniline (40 g, 233 mmol) in 1,4-dioxane (480 mL) was added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (94 g, 372 mmol), ium acetate (45.6 g, 465 mmol), 2-dicyclohexylphosphino-2',4',6'-tri-i-propyl-1,1'-biphenyl (X-phos) (8.07 g, 13.95 mmol), tris(dibenzylideneacetone)dipalladium(0) (8.52 g, 9.30 mmol). Then the mixture was heated at 80°C for 4 hours under nitrogen. Another additional vial was set up as described above. Two reaction mixtures were combined, concentrated and the residue was purified by column chromatography on silica gel (eluted with PE/EtOAc = 10/1) to obtain the target compound (60 g, yield 55.4%) as light yellow solid. 1H NMR (400MHz, chloroform-d) 8 7.23 -7.13 (m, 3H), 6.80 (d, J=7.5 Hz, lH), 3.82 -3.38 (m, 2H), 1.34 (s, 12H).
Step 3: Synthesis of tert-butyl (3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolanyl)phenyl) p-s:+ 3-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolanyl)aniline (30 g, 137 mmol) and di-tert-butyl dicarbonate (38.9 g, 178 mmol) were mixed in e (600 mL) at 100°C for 24 hours. Another additional vial was set up as described above. Two reaction mixtures were combined. The brown mixture was evaporated, dissolved in EtOAc (1.5 L), washed with 0.1 N HCl (3 X 2 L) and brine (3 L), dried over Na2SO4, filtered and trated under reduced pressure to give the title compound (50 g, yield 57%) as red solid. 1H NMR (400MHz, chloroform-d) 8 7.63 (br. s., 2H), 7.48 (d, J=7.l Hz, lH), 7.37 -7.28 (m, lH), 1.52 (s, 9H), 1.34 (s, 12H).
Step 4: Synthesis oftert-butyl (3-(4-formylbenzyl)phenyl)carbamate F\.-s._ O- 'J-fl o-\- + I - HN ✓,; Br A mixture of 4-(bromomethyl)benzaldehyde (24.94 g, 125 mmol), 1,1'- phenylphosphino) ferrocenedichloro palladium(II) DCM complex (13.75 g, 18.80 mmol), tert-butyl 4,5,5-tetramethyl- l,3,2-dioxaborolanyl)phenyl)carbamate (20 g, 62.7 mmol) and potassium carbonate (43.3 g, 313 mmol) in tetrahydrofuran (400 mL) was heated to 80°C for 12 hours. Another additional vial was set up as described above. Two reaction mixtures were combined. The on mixture was diluted with water (500 mL). The aqueous layer was extracted with EtOac (3 x 500 mL).
The organic layer was dried over Na2S04, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (eluted with Ac = 10/1) to obtain the title compound (15 g, yield 38.4%) as white solid. 1H NMR (400MHz, form-d) 8 9.95 (s, lH), 7.78 (d, J=7.9 Hz, 2H), 7.33 (d, J=7.9 Hz, 2H), 7.27 - 7.13 (m, 3H), 6.82 (d, J=7.l Hz, lH), 6.47 (br. s., lH), 4.00 (s, 2H), 1.48 (s, 9H).
Step 5: Synthesis of (6S,8S,9R,10S,11S,13S,14S,16R, l7S)-6,9-difluor o-l l,16, l 7- trihydroxy(2-hydroxyacetyl)- l0,13-dimethyl-6,7,8,9,10, 11,12,13,14, 15,16, l7-dodecahydro-3H­ cyclopenta[ a]phenanthrenone OH OH i= i= (2S,6aS,6bR,7S,8aS,8bS, l laR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)- 6a,8a,10,10-tetramethyl- l,2,6a,6b,7,8,8a,8b, l 12a,12b-dodecahydro-4H- naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone (20 g, 44.2 mmol) was suspended in 40% aqueous HBF4 (440 mL) and the mixture was stirred at 25°C for 48 hours. After the reaction was complete, 2 L ofH20 was added and the solid was collected by filtration to give a white solid. This solid was washed with H20 (1 L) and then MeOH (200 mL) to give the title compound (11 g, yield 60.3%) as a white solid. 1H NMR z, DMSO-d6) 8 7.25 (d, J=l0. l Hz, lH), 6.28 (d, J=l0.l Hz, lH), 6.10 (s, lH), 5.73 -5.50 (m, lH), 5.39 (br. s., lH), 4.85 -4.60 (m, 2H), 4.50 (d, J=l9.4 Hz, lH), 4.20 - 4.04 (m, 2H), 2.46 - 2.06 (m, 6H), 1.87 -1.75 (m, lH), 1.56 - 1.30 (m, 6H), 0.83 (s, 3H).
Step 6: Synthesis of (2S,6aS,6bR,7S,8aS,8bS, l0S, l laR,12aS,12bS)- l 0-( 4-(3- aminobenzyl)phenyl)-2,6b-difluo rohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone ] A suspension of (6S,8S,9R,10S,11S,13S,14S,16R,17S)-6,9-difluoro-l l,16,17-trihydroxy-l 7- (2-hydroxyacetyl)-l0,13-dimethyl-6,7,8,9,10,11, 12, 13,14,15, 16,l7-dodecahydro-3H- cyclopenta[a]phenanthrenone (4.4 g, 10.67 mmol) and magnesium sulfate (6.42 g, 53.3 mmol) in MeCN (100 mL) was allowed to stirred at 20°C for 1 hour A solution of tert-butyl (3-(4- formylbenzyl)phenyl)carbamate (3.65 g, 11.74 mmol) in MeCN (100 mL) was added in one portion.
Trifluoromethanesulfonic acid (9.01 mL, 53.3 mmol) was added drop wise while maintaining an internal temperature below 25°C using an ice bath. After the addition, the mixture was stirred at 20°C for 2 hours.
Three additional vials were set up as described above. All four reaction mixtures were combined. The mixture solution was trated and the residue was purification by Prep-HPLC to give the title compound (4.5 g, yield 14.2%) as yellow solid. LCMS (Method b, Table 7) R1 = 2.65 min; MS m/z = 606.2 (M+H)\ 1HNMR (400MHz, DMSO-d6) 8 7.44 - 7.17 (m, 5H), 6.89 (t, J=7.7 Hz, lH), 6.44 -6.25 (m, 4H), 6.13 (br. s., lH), 5.79 - 5.52 (m, 2H), 5.44 (s, lH), 5.17 - 4.89 (m, 3H), 4.51 (d, J=l9.4 Hz, lH), 4.25 - 4.05 (m, 2H), 3.73 (s, 2H), 3.17 (br. s., lH), 2.75 - 2.55 (m, lH), 2.36 - 1.97 (m, 3H), 1.76 -1.64 (m, 3H), 1.59 - 1.39 (m, 4H), 0.94 - 0.78 (m, 3H). Prep-HPLC Method: Instrument: Gilson 281 semipreparative HPLC system; Mobile phase: A: Formic Acid/H2O=0.0l % v/v; B: CH3CN; Column: Luna Cl8150*25 5 micron; Flow rate: 25 mL/min; r wavelength: 220 and 254nm.
Time 0.0 10.5 10.6 10.7 13.7 13.8 15.0 B% 15 35 35 100 100 10 10 Example 2A: Synthesis of (6aR,6bS,7S,8aS,8bS,1 OR, l laR,12aS,12bS)-l 0-(4-(3-aminobenzyl)phenyl)- oxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl-l,2,6a,6b,7,8,8a,8b,l la,12,12a,12b-dodecahydro-4H­ o[2', l indeno[l,2-d][ l,3]dioxolone (Cpd. No. 41) Step 1: Synthesis of 4-(bromomethyl)benzaldehyde NC� --------1► OHC� �Br �Br To a solution of 4-(bromomethyl)benzonitrile (50 g, 255 mmol) in toluene (1 L) was added diisobutylaluminum hydride (383 mL, 383 mmol, 1 M in toluene) dropwise at 0°C. The mixture was stirred for 1 hour Two additional vials were set up as described above. All three reaction es were combined. 10% aqueous HCl (1.5 L) was added and then extracted with DCM (3 X 1.5 L). The organic layer was dried over Na2SO4, ed and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel d with petroleum ether/ethyl acetate 10/1) to afford the title nd (120 g, 82%) as a white solid. 1H NMR (400MHz, CDC1 3) 8 10.01 (s, lH), 7.86 (d, J 8.4 Hz, 2H), 7.55 (d, J=7.9 Hz, 2H), 4.51 (s, 2H).
Step 2: Synthesis of 3-( 4,4,5,5-tetramethyl-l,3,2-dioxaborolanyl)aniline To a solution of 3-bromoaniline (80 g, 465 mmol) in 1,4-dioxane (960 mL) was added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (177 g, 698 mmol), potassium acetate (91 g, 930 mmol), 2-dicyclohexylphosphino-2',4',6'-tri-i-propyl-l,l'-biphenyl (13.45 g, 23.25 mmol) and tris(dibenzylideneacetone)dipalladium(0) (17.03 g, 18.60 mmol). The mixture was heated at 80°C for 4 hours under nitrogen. Two additional vials were set up as described above. Three reaction es were combined, concentrated and the residue purified by column chromatography on silica gel ( eluted with petroleum ethyl acetate 10/1) to afford the title compound (150 g, 46.6 %) as a light yellow solid. 1H NMR (400MHz, CDCh) 8 7.23 - 7.13 (m, 3H), 6.80 (d, J=7.5 Hz, lH), 3.82 -3.38 (m, 2H), 1.34 (s, 12H).
Step 3: Synthesis of tert-butyl (3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan yl)phenyl)carbamate o, .,,o Q ,Boc 3-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolanyl)aniline (50 g, 228 mmol) and di-tert-butyl dicarbonate (64.8 g, 297 mmol) were mixed in toluene (500 mL) and the mixture stirred at 100°C for 24 hours. Two onal vials were set up as described above. The three reaction mixtures were combined.
The brown mixture was concentrated and the residue was washed with PE to afford the title compound (120 g, 49.5 %) as a white solid. 1H NMR (400MHz, CDCh) 8 7.62 (s, 2H), 7.48 (d, J=7.5 Hz, lH), 7.35 - 7.29 (m, lH), 6.46 (br. s., lH), 1.52 (s, 9H), 1.34 (s, 12H).
Step 4: Synthesis -butyl (3-(4-formylbe nzyl)phenyl)carbamate �Br ;to Boe I I N ..-B - � 'Boc 0 NH OHC� yy 0 OHC� � A mixture of 4-(bromomethyl)benzaldehyde (29.9 g, 150 mmol), 1,1'-bis(diphenylphosphino) ferrocenedichloro palladium(II) (20.63 g, 28.2 mmol), tert-butyl (3-(4,4,5,5-tetramethyl- l,3,2- dioxaborolanyl)phenyl)carbamate (30 g, 94 mmol) and potassium carbonate (64.9 g, 470 mmol) in THF (600 mL) was heated to 80°C for 12 hours. Three additional vials were set up as described above.
All four on mixtures were combined. The on mixture was diluted with water (1 L). The aqueous layer was extracted with EtOAc (3 X 800 mL). The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (eluted with PE/EtOAc =10/1) to afford the title compound (35.5 g, 27.3 %) as a white solid. 1H NMR (400MHz, CDCh) 8 9.97 (s, lH), 7.80 (d, J=7.9 Hz, 2H), 7.35 (d, J=7.9 Hz, 2H), 7.26 (s, 2H), 7.24 - 7.13 (m, 2H), 6.84 (d, J=7.l Hz, lH), 6.43 (br. s., lH), 4.02 (s, 2H), 1.50 (s, 9H).
Step 5: Synthesis of (6aR,6bS,7S,8aS,8bS,l0R,llaR,12aS,12bS)(4-(3- aminobenzyl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll 6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxolone To a solution of (8S,9S,10R,11S,13S,14S,16R,17S)-ll,16,17-trihydroxy- l 7-(2- hydroxyacetyl)-l 0,13-dimethyl-6,7,8,9,10, 11, 12,13,14, 15, 16, l7-dodecahydro-3H- cyclopenta[a]phenanthrenone (6 g, 15.94 mmol) and tert-butyl (3-(4-formylbenzyl)phenyl)carbamate (4.96 g, 15.94 mmol) in MeCN (50 mL) was added perchloric acid (4.79 mL, 80 mmol) dropwise while ining an internal temperature below 25°C using an ice bath. After the addition, the e was stirred at 20°C for 2 hours. Three additional vials were set up as described above. All four reaction mixtures were ed. The reaction mixture was quenched with saturated NaHCO 3 aqueous (500 mL) and extracted with dichloromethane (3 X 800 mL). The c phase was concentrated and the residue was purified by Prep-HPLC to afford the title compound (10 g, 27.0 %) as a yellow solid. 1H NMR (400MHz, DMSO-d6) 8 7.36 (d, J=7.9 Hz, 2H), 7.31 (d, J=l0.l Hz, lH), 7.20 (d, J=7.9 Hz, 2H), 6.89 (t, J=7.9 Hz, lH), 6.39 -6.28 (m, 3H), 6.16 (dd, J=l.5, 9.9 Hz, lH), 5.93 (s, lH), 5.39 (s, lH), 5.08 (t, J=5.7 Hz, lH), 4.98 -4.87 (m, 3H), 4.78 (d, J=3.l Hz, lH), 4.49 (dd, J=6.2, 19.4 Hz, lH), 4.29 (br. s., lH), 4.17 (dd, J=5.5, 19.6 Hz, lH), 3.74 (s, 2H), 2.61 -2.53 (m, lH), 2.36 - 2.26 (m, lH), 2.11 (d, J=l l.0 Hz, lH), 2.07 (s, lH), 2.02 (d, J=l2.8 Hz, lH), 1.83 - 1.54 (m, 5H), 1.39 (s, 3H), 1.16 -0.96 (m, 2H), 0.85 (s, 3H).
LCMS: t =2.365 min, 98% purity, m/z =570.2 (M+Ht LC/MS (Table 7, method a) Method of Prep-HPLC: Instrument: Gilson 281 semi-preparative HPLC system, Mobile phase: A: CF3COOH/H2O=0.075% v/v; B: CH3CN, Column: Phenomenex Luna(2) Cl8 250*50 l0u, Flow rate: 80 mL/min, Monitor wavelength: 220&254 nm, Time B%, 0.0 28, 20.0 45, 20.1 45, .2 100, 30.2 100, 0.3 28, 31.5 28.
Example 2B: Synthesis of(2R,6aS,6bR,7S,8aS,8bS, lOR, llaR,12aS,12bS)(4-(3-Aminobenzyl)phenyl)-6bfluoro-2 ,7-dihydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl-6a,6b, 7,8,8a,8b, lla, 12,12a, 12bdecahydro-1H-naphtho [2',l':4,5]indeno[1,2-d][1,3]dioxol-4(2H)-one :: H 0- •'""'v-' N y •OH + � 'Boc _ 10 NH2 OHCA:s} V 0 0 OH OH Trifluoromethane sulfonic acid (1.34 ml, 15.11 mmol) was added drop-wise to a -10 °C suspens10n of (6R,8S,9R,10S,11S,13S,14S,16R,l 7S)fluoro-6,l l,16,17-tetrahydroxy- l7-(2- hydroxyacetyl)-l0,13-dimethyl-6,7,8,9, 10,11,12,13,14, 15, 16, l7-dodecahydro-3H- cyclopenta[a]phenanthrenone (1.55g, 3 .78 mmol), tert-butyl (3-(4-formylbenzyl)phenyl)carbamate (from Example 2, step 4) (1.176 g, 3.78 mmol), and MgSO4 (2.273 g, 18.89 mmol) in MeCN (15.1 mL).
After 20min, the reaction was quenched by addition of a saturated aqueous solution ofNaHCO 3 (15 mL), ed by water (60 mL) and EtOAc (l00mL). The organic layer was washed sequentially with water (60 mL), brine (60 mL), dried (Na 2SO4), and t was removed under reduced pressure. Purification by chromatography (silica, 40 g) eluting with a gradient of40-100% heptanes provided the title compound as a foam (880 mg, 1.458 mmol, 39% yield) in 90% . The product could be further purified by reverse phase HPLC on a Waters XBridge™ RP18 5 micron column (30 x 100 mm). A nt ofMeCN (A) and 0.1 mM NH4CO3 in water (B) was used, at a flow rate of40 mL/min (0-5.0 min 5% A, 5.0-19.0 min linear gradient 15-55% A). LC-MS (Method r, Table 7) Rt 0.72 min, m/z 604.3 [M+H+]. 1H NMR (500 MHz, DMSO-d 10.1 Hz, lH), 7.24 - 6) 8 7.37-7.31 (m, 2H), 7.28 (d, J 7.19 (m, 2H), 6.93 -6.85 (m, lH), 6.36 (d, J 2.1 Hz, 2H), 6.35 (p, J 1.1 Hz, lH), 6.23 (dd, J 10.1, 1.9 Hz, lH), 6.10 (d, J 1.9 Hz, lH), 5.45 (s, lH), 5.38 (s, lH), 5.10 (s, lH), 4.96 -4.91 (m, 3H), 4.51 (d, J= 19.4 Hz, lH), 4.38 (s, lH), .16 (m, 2H), 3.74 (s, 2H), 2.76-2.60 (m, lH), 2.20 (td, J= 12.5, 6.3 Hz, lH), 2.08 (s, 2H), 1.86 (d, J 11.8 Hz, lH), 1.75-1.58 (m, 7H), 0.89 (s, 3H).
Example 3: Synthesis of bR,7S,8aS,8bS,l0R, l laR,12aS,12bS)(4-((3- aminophenyl)thio)phenyl)-6b-fluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[ l,2-d][l,3]dioxolone Step 1: Synthesis oftert-butyl (3-mercaptophenyl)carbamate To a mixture of zinc perchlorate (0.422 g, 1.598 mmol) and obenzenethiol (10 g, 80 mmol) was added di-tert-butyl dicarbonate (22.66 g, 104 mmol) drop wise. The solution was stirred at °C for 12 hours. Three additional vials were set up as described above. Four reaction mixtures were combined. The mixture was ved in EtOAc (200 mL) and washed with water (500 mL). The organic layer was dried over Na2S04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography on silica gel (eluted with Ac = 5/1) to obtain the target compound (50 g, yield 69 .4 %) as white solid. 1H NMR ( 400 MHz, CDC1 3) 8 7.45 (br. s., lH), 7.16 - 7.09 (m, lH), 7.06 - 7.01 (m, lH), 6.92 (d, J 7.4Hz, lH), 6.55 (br. s., lH), 3.46 (s, lH), 1.52 (s, 9H).
Step 2: Synthesis of tert-butyl (3-((4-formylphenyl)thio)phenyl)carbamate � r(YCHO Boc,N�S� To a solution of utyl (3-((4-formylphenyl)thio)phenyl)carbamate (10 g, 44.4 mmol) in DMF (300 mL) was added triphenylphosphine (11.64 g, 44.4 mmol) and N-ethyl-N-isopropylpropan amine (11.47 g, 89 mmol) at 25°C. The reaction mixture was stirred at 25°C for 30 minutes under N2 .
The mixture was added 4-fluorobenzaldehyde (8.26 g, 66.6 mmol) at 100°C and the mixture was stirred at 100°C for 12 hours. Four additional vials were set up as described above. The five reaction mixtures were combined. The mixture was d with water (2 L) and ted with EtOAc (3 x 1 L). The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (eluted with PE/EtOAc = 10/1) to obtain the target compound (55 g, yield 75%) as yellow oil. 1HNMR (400 MHz, CDCh) 8 9.90 (s, lH), 7.71 (d, J=8.4 Hz, 2H), 7.58 (s, lH), 7.48 - 7.41 (m, lH), 7.33 (t, J 7.9 Hz, lH), 7.25 (d, J=8.4 Hz, 2H), 7.17 (d, J=7.9 Hz, lH), 6.72 (br. s., lH), 1.50 (s, 9H).45 (br. s., lH), 7.16 -7.09 (m, lH), 7.06 - 7.01 (m, lH), 6.92 (d, J 7.4 Hz, lH), 6.55 (br. s., lH), 3.46 (s, lH), 1.52 (s, 9H).
Step 3: Synthesis of sodium (4-((3-((tert-butoxycarbonyl)amino)phenyl)thio)phenyl) (hydroxy)methanesulfonate To a solution of the aldehyde (15 g, 45.5 mmol) in CH3CN (30 mL) was added a solution of sodium metabisulfite (11.25 g, 59.2 mmol) in water (90 mL) at 25°C. The mixture was stirred at 25°C for 48 hours. Another additional vial was set up as described above. Two reaction mixtures were ed.
The on was ed and the solid was washed with water (150 mL), CH3CN (150 mL) and dried under reduced pressure to give the target compound (32 g, yield 81%) as white solid.1H NMR ( 400MHz, DMSO-d6) 8 9.45 (br. s., lH), 7.54 - 7.49 (m, lH), 7.47 -7.35 (m, 3H), 7.33 -7.17 (m, 3H), 6.85 (d, J=7.9 Hz, lH), 5.97 (d, J 4.9 Hz, lH), 4.98 (d, J 4.9 Hz, lH), 1.45 (s, 9H).
Step 4: Synthesis of (6aS,6bR,7S,8aS,8bS,l0R)laR,12aS,12bS)(4-((3- aminophenyl)thio)phenyl)-6b-fluorohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxolone To a solution of (8S,9R)0S,11S,13S,14S,16R)7S)fluoro-ll,16, l7-trihydroxy-l7-(2- hydroxyacetyl)-10,13-dimethyl-6,7,8,9,10,11,12,13,14, 15, 16, ecahydro-3H- cyclopenta[a]phenanthrenone (6 g, 15.21 mmol) and sodium (4-((3-((tertbutoxycarbonyl )amino)phenyl)thio)phenyl)(hydroxy)methanesulfonate (4.74 g, 15.21 mmol) in THF (50 mL) was added perchloric acid (4.58 mL, 76 mmol) drop wise while maintaining an internal ature below 25°C using an ice bath. After the addition, the e was stirred at 20°C for 2 hours. Three additional vials were set up as described above. All four reaction es were combined. The reaction mixture was quenched with sat. NaHCO3 aqueous (500 mL) and extracted with DCM (3 X 800 mL). The organic phase was concentrated and the residue was purification by Prep-HPLC to give the target compound (9.5 g, 25.8 %) as yellow solid. LCMS (Method b, Table 7) R1 = 2.68 min, m/z = 588.1 (M+H)\ 1H NMR z, DMSO-d6) 8 7.37 - 7.26 (m, 3H), 7.21 (d, J=7.9 Hz, 2H), 6.89 (t, J=7.7 Hz, lH), 6.43 -6.30 (m, 3H), 6.23 (d, J=l0.l Hz, lH), 6.04 (s, lH), 5.75 (s, lH), 5.44 (s, 2H), 5.09 (t, J=5.7 Hz, lH), 4.93 (br. s., 3H), 4.50 (dd, J=6.2, 19.4 Hz, lH), 4.28 - 4.09 (m, 2H), 3.74 (s, 2H), 2.73 -2.54 (m, 2H), 2.35 (d, J=l3.2 Hz, lH), 2.25 - 2.12 (m, lH), 2.05 (d, J=l5.0 Hz, lH), 1.92 - 1.77 (m, lH), 1.74 - 1.58 (m, 3H), 1.50 (s, 3H), 1.45 -1.30 (m, lH), 0.87 (s, 3H). Prep-HPLC Method: Instrument: Gilson 281 semi-preparative HPLC system; Mobile phase: A: CF3CO2H/H2O=0.075% v/v; B: CH3CN; Column: Phenomenex Luna Cl8 250 x *50mm*10 micron; Flow rate: 80mL/min; Monitor wavelength: 220 and 254nm.
Time 0.0 20.0 20.1 20.2 30.2 30.3 31.5 B% 10 42 42 100 100 10 10 Example 4: Synthesis of (6aR,6bS,7S,8aS,8bS, l0R l laR12aS,12bS)(4-((3- aminophenyl)thio)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b, l 12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxolone To a solution of (8S,9S,10R,11S,13S,14S,16R,17S)-l l,16,17-trihydroxy- l7-(2- hydroxyacetyl)-10,13-dimethyl-6,7,8,9,10, 11,12,13,14,15,16, l7-dodecahydro-3H- cyclopenta[a]phenanthrenone (6 g, 15.94 mmol) and sodium (4-((3-((tert- butoxycarbonyl)amino)phenyl)thio)phenyl)(hydroxy)methanesulfonate (4.96 g, 15.94 mmol) in MeCN (50 mL) was added perchloric acid (4.79 mL, 80 mmol) drop wise while maintaining an internal temperature below 25°C using an ice bath. After the addition, the mixture was stirred at 20°C for 2 hours.
Three additional vials were set up as described above. All four on mixtures were combined. The reaction mixture was quenched with sat. s NaHCO3 (500 mL) and extracted with DCM (3 X 800 mL). The organic phase was concentrated and the residue was purification by Prep-HPLC to give the target compound (10 g, 27.0 %) as yellow solid. LCMS (Method b, Table 7) R1 = 2.36 min, m/z = 570.2 (M+H)\ 1H NMR (400MHz, DMSO-d6) 8 7.36 (d, J=7.9 Hz, 2H), 7.31 (d, J=l0.l Hz, lH), 7.20 (d, J=7.9 Hz, 2H), 6.89 (t, J=7.9 Hz, lH), 6.39 - 6.28 (m, 3H), 6.16 (dd, J=l.5, 9.9 Hz, lH), 5.93 (s, lH), 5.39 (s, lH), 5.08 (t, J=5.7 Hz, lH), 4.98 - 4.87 (m, 3H), 4.78 (d, J=3. l Hz, lH), 4.49 (dd, J=6.2, 19.4 Hz, lH), 4.29 (br. s., lH), 4.17 (dd, J=5.5, 19.6 Hz, lH), 3.74 (s, 2H), 2.61 - 2.53 (m, lH), 2.36 - 2.26 (m, lH), 2.11 (d, J=l l.0 Hz, lH), 2.07 (s, lH), 2.02 (d, J=l2.8 Hz, lH), 1.83 - 1.54 (m, 5H), 1.39 (s, 3H), 1.16 - 0.96 (m, 2H ), 0.85 (s, 3H ). PLC Method: Instrument: Gilson 281 semi-preparative HPLC system; Mobile phase: A: CF3CO2H/H2O=0.075% v/v; B: CH3CN; Column: Phenomenex Luna Cl8 250 x *50 mm*10 ; Flow rate: 80mL/min; Monitor wavelength: 220 and 254nm.
Time 0.0 20.0 20.1 20.2 30.2 30.3 31.5 B% 28 45 45 100 100 28 28 Example 5: Synthesis of(2S,6aS,6bR,7S,8aS,8bS,1 OR l laR12aS,12bS)-l0-(4-((3-aminophenyl) thio)phenyl)-2, 6b-difluorohydroxy-8b-(2-hydroxyacety 8a-dimethyll ,2,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[ [l,3]dioxolone E E To a solution of steroid (10 g, 24.25 mmol) in CH3CN (200 mL) was added ium sulfate (10.21 g, 85 mmol) at 25°C. The mixture was stirred at 25°C for 4 hours. Then to the above solution was added sodium ( 4-( (3-((tert-butoxycarbonyl)amino)phenyl)thio) )(hydroxy)methanesulfonate (10.51 g, 24.25 mmol) and oromethanesulfon ic acid (20.48 mL, 121 mmol) at 0°C. The resulting mixture was stirred at 25°C for 1 hour. Two additional vials were set up as described above. Three reaction mixtures were combined. The mixture was diluted with 1 N NaOH (300 mL) and extracted with EtOAc (3 x 600 mL). The organic layer was concentrated under reduced pressure to give a residue. The residue was dissolve in EtOAc (60 mL) and added 2-butanone (180 mL). After stirring for 30 minutes, the solid was collected by filtration and purified by Prep-HPLC to give the title compound (8.4 g, yield 17.52 %) as yellow solid. LCMS (Method c, Table 7) R1 = 2.66 min; MS m/z = 624.1 (M+H)\ 1H NMR (400MHz, 6) 8 7.39 (d, J=8.4 Hz, 2H), 7.25 (d, J=8.4 Hz, 3H), 7.03 (t, J=7.7 Hz, lH), 6.61 (s, lH), 6.53 (t, J=8.2 Hz, 2H), 6.29 (dd, J=l.5, 9.9 Hz, lH), 6.12 (s, lH), 5.76 - 5.49 (m, 2H), 5.46 (s, lH), 4.96 (d, J=4.9 Hz, lH), 4.52 (d, J=l9.4 Hz, lH), 4.21 (d, J=l9.4 Hz, 2H), 2.74 - 2.53 (m, 2H), 2.34 -2.13 (m, 2H), 2.09 - 1.96 (m, lH), 1.79 -1.62 (m, 3H), 1.57 - 1.43 (m, 4H), 0.86 (s, 3H). Prep-HPLC method: Instrument: Shimadzu LC-8A preparative HPLC; Column: Phenomenex Luna Cl8 250 x *50mm*10 micron; Mobile phase: A for H2O (0.09% CF3CO2H) and B for CH3CN; Gradient: B from 22% to 52% in 20 min; Flow rate: 80 mL/min; Wavelength: 220&254 nm. e 6: Synthesis of ( 6aR6bS, 7S,8aS,8bS, 1 OR, l laR,12aS,12bS)- l0-(4-(3-Amino hydroxybenzyl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl -6a,6b,7,8,8a,8b, l la, 12,12a,12bdecahydro-1H-naphtho [2', l ': 4,5]indeno[ 1,2-d][ l ,3]dioxol-4(2H)-one HO ov-OH TfOH, MgSO4 ACN, 0 5 °C 0 .. ,., 7 ,,1\\0 Triflic acid (0.2 mL, 2.183 mmol) was added drop-wise to a 0 °C slurry of (8S,9S,10R,l IS,13S,14S,16R,17S)-l l,16,17-trihydroxy(2-hydroxyacetyl)-10,13-dimethyl- 6,7,8,9,10,l 3,14,15,16, ecahydro-3H-cyclopenta[a]phenanthrenone (0.164 g, 0.437 mmol), tert-butyl (2-((tert-butyldimethylsilyl)oxy)(4-formylbenzyl)phenyl)carbamate (0.193 g, 0.437 mmol) and MgSO4 (0.189 g, 1.572 mmol) in MeCN (1.8 mL). After 40 min the reaction was d with EtOAc (15 mL), and then washed sequentially with a ted aqueous solution ofNaHCO3 (10 mL x 2), and with a saturated aqueous solution of brine (5 mL). The organic phase was dried (Na 2SO4) and solvent was removed under reduced pressure. Purification by tography (silica, 12 g) g with a gradient of 0-10% MeOH/DCM gave the title compound (163 mg, 0.278 mmol, 64 % yield) as a waxy solid. A portion of this material (ca. 48.9 mg) was further purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 X 50 mm column). A gradient ofMeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of90 mL/min (0-5.0 min 15% A, 5.0-20 min linear gradient 15-70% A, hold 2 min). Combined fractions were concentrated under reduced pressure to remove volatile solvents, and the resulting solution was frozen and lyophilized to give a off-white solid (11.9 mg). LCMS (Method r, Table 7) R1 0.75 min, m/z 586.26 [M+H+]. 1H NMR (400 MHz, DMSO-d6) 8 10.27 (s, lH), 9.04 (s, 2H), 7.34 (d, J= 8.0 Hz, 2H), 7.28 (d, J= 10.1 Hz, lH), 7.18 (d, J= 8.0 Hz, 2H), 6.94 (dd, J= 8.1, 2.1 Hz, lH), 6.90 (d, J 2.1 Hz, lH), 6.82 (d, J 8.2 Hz, lH), 6.17 - 6.07 (m, lH), 5.90 (d, J 1.6 Hz, lH), 5.37 (s, lH), 4.89 (d, J 4.9 Hz, lH), 4.75 (s, lH), 4.46 (d, J 19.4 Hz, lH), 4.26 (q, J 3.3 Hz, lH), 4.14 (d, J 19.5 Hz, lH), 3.80 (s, 2H), 2.58 - 2.46 (m, lH), 2.36 - 1.92 (m, 3H), 1.76 -1.56 (m, 4H), 1.36 (s, 3H), 1.10 - 0.90 (m, 2H), 0.83 (s, 3H).
Example 7: Synthesis of(2S,6aS,6bR7S,8aS,8bS, 1 OR l laR,12aS,12bS)-l 0-( 4-(3-Aminobenzyl) hydroxyphenyl)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl - 6a,6b,7,8,8a,8b, l la,12,12a,12b-decahydro-1H-naphtho[2',l ':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one 0] Step 1: Synthesis of4-((3-bromophenyl)(hydroxy)methyl)methoxybenzonitrile i-PrMgCl•LiCI Br THF, then M m-BrPhCHO �Br NC�OMe NCJl) V Isopropylmagnesium chloride lithium chloride complex (1.3 M in THF, 8.34 mL, 10.85 mmol) was added drop-wise to a 0-5 °C solution of4-bromomethoxybenzonitrile (2 g, 9.43 mmol) in THF (21 mL). The on was stirred for 5h, whereupon a solution of3-bromobenzaldehyde (1.979 g, .38 mmol) in THF (10.5 mL) was added drop-wise, maintaining a temperature of <10°C. The reaction was permitted to slowly warm to room temperature overnight. The reaction quenched with a saturated aqueous on ofNH4Cl (25 mL) and extracted with MTBE (50 mL X 3). The ed organics were washed with brine (20 mL), dried (Na 2SO4), and solvents were removed under reduced pressure.
Purification by chromatography (80 g silica) eluting with a gradient of 0-10% MTBE/heptanes gave the title nd (1.77 g, 5.56 mmol, 59 % yield) as a beige syrup/oil. LCMS (Method r, Table 7) Rt 0.86 min; MS (ESI-) m/z 315.7 [M-H+]. 1H NMR (501 MHz, DMSO-d6) 8 7.67 (d, J 7.8 Hz, lH), 7.47 (t, J 1.8 Hz, lH), 7.43 (dd, J 7.8, 1.5 Hz, lH), 7.41 (d, J l.4 Hz, lH), 7.40 - 7.36 (m, lH), 7.28 (dt, J 7.8, 1.5 Hz, lH), 7.23 (d, J 7.8 Hz, lH), 6.10 (d, J 4.4 Hz, lH), 5.94 (d, J 4.1 Hz, lH), 3.80 (s, 3H).
Step 2: Synthesis of4-(3-bromobenzyl)methoxybenzonitrile M Nal, TMSCI �Br ACN, 55 °C �Br NC)__)V V Chlorotrimethylsilane (3.63 g, 33.4 mmol) was added to a room temperature solution of sodium iodide (5.00 g, 33.4 mmol) in MeCN (18.5 mL), which resulted in the ate precipitation of a white solid. A solution of 4-((3-bromophenyl)(hydroxy)methyl)methoxybenzonitrile (1.77 g, 5.56 mmol) in MeCN (18.5 mL) was then added, whereupon the reaction mixture was heated to 55 °C for 60 min. After cooling to room temperature the reaction was partitioned n MTBE (50 mL) and water (50 mL). After separating the layers the s phase was extracted with MTBE (50 mL X 2). The combined organics were washed tially with a 1 M aqueous solution of Na2S2O3 (50 mL X 2), followed by a saturated aqueous solution of brine (30 mL), dried (Na 2SO4), and solvents were removed under reduced pressure. cation by chromatography (silica, 80 g) eluting with a gradient of 5-40% MTBE/heptanes gave the title compound (1.58 g, 5.23 mmol, 94% yield) as an off-white solid. LCMS (Method r, Table 7) Rt 1.02 min; MS m/z not observed. 1H NMR (501 MHz, DMSO-d6) 8 7.42 (d, J 1.5 Hz, lH), 7.39 - 7.30 (m, 4H), 7.22 (td, J 7.6, 0.6 Hz, lH), 7.18 (dt, J 7.7, 1.4 Hz, lH), 3.94 (s, 2H), 3.82 (s, 3H).
Step 3: Synthesis of4-(3-bromobenzyl)methoxybenzaldehyde OMe DiBAI-H �Br /hen1Naq.HCI �Br NC)l)V OHC)l)V Diisobutylaluminum hydride (4.9 mL, 1.0 M solution in hexanes, 4.9 mmol) was added dropwise over 5 min to a O °C solution of 4-(3-bromobenzyl)methoxybenzonitrile (0.99 g, 3.28 mmol) in toluene (16 mL), maintaining a temperature of< 6 °C. After 10 min the reaction was quenched by careful addition of a 1 N aqueous solution ofHCl (100 mL) at O °C. It was then extracted with DCM (50 mL X 4), washed with a saturated s solution of brine (30 mL), and solvent was removed under reduced pressure. Purification by chromatography (silica, 40 g) eluting with a gradient of0-40% MTBE/heptanes gave the title compound (780 mg, 2.56 mmol, 78% yield) as a ess oil. LCMS (Method r, Table 7) Rt 0.95 min, MS (DCI+) m/z 303.9, 305.9 (M+). 1H NMR (400 MHz, DMSO-d6) 8 9.93 (s, lH), 7.47 (dd, J 7.5, 1.5 Hz, lH), 7.44 (d, J 1.5 Hz, lH), 7.42 - 7.33 (m, 3H), 7.25 -7.17 (m, 2H), 3.96 (s, 2H), 3.85 (s, 3H). 6] Step 4: Synthesis of4-(3-bromobenzyl)hydroxybenzaldehyde g g_� �Br ) OHC)l_) V Boron tribromide (1.0 Min methylene chloride, 6.4 mL, 6.4 mmol) was added drop-wise to a 0-3 °C solution of 4-(3-bromobenzyl)methoxybenzaldehyde (0.78 g, 2.56 mmol) in DCM (7.8 mL).
The reaction was stirred at O °C for 30 min; then was stirred for 90 min at room temperature. t was removed under reduced pressure and the resulting dark oil was treated with MeOH (20 mL) and water (15 mL), which gave a heterogeneous mixture. MeCN was added until a homogeneous solution was obtained (ca. 10 mL) and the solution was stirred overnight. Volatile solvents were removed under reduced pressure and the resulting aqueous suspension was extracted with DCM (25 mL X 3). The combined cs were washed with brine (20 mL), dried over Na2S04 and solvent was removed under reduced pressure. Purification by chromatography (silica, 40 g) eluting with a gradient of10-50% eptanes gave 4-(3-bromobenzyl)hydroxybenzaldehyde (660 mg, 2.267 mmol, 89% yield) as a white solid.
LCMS (Method r, Table 7) Rt 0.85 min; MS (DCI+) m/z , 309.97 [M+NH/]. 1H NMR (400 MHz, DMSO-d6) 8 10.07 (s, lH), 9.83 (s, lH), 7.39 (q, J 1.3 Hz, lH), 7.33 (ddt, J 6.5, 4.4, 2.0 Hz, lH), 7.30 (d, J 0.9 Hz, 2H), 7.25 (s, lH), 7.25 - 7.15 (m, 2H), 3.92 (s, 2H).
Step 5: Synthesis of4-(3-bromobenzyl)((tert-butyldimethylsilyl)oxy)benzaldehyde OH TBSCI �Br imid., DCM �Br OHC)l_) V ) V Imidazole (0.231 g, 3.40 mmol) and tert-butyldimethylchlorosilane (0.410 g, 2.72 mmol) were added to a room ature suspension of 4-(3-bromobenzyl)hydroxybenzaldehyde (0.660 g, 2.267 mmol) in DCM (7.6 mL), which was d for 3 h. MeOH (0.5 mL) was added and stirring continued for 10 min, whereupon the reaction was diluted with DCM (100 mL), washed sequentially with water (25 mL), a 1 N aqueous solution ofHCl (25 mL), and with a saturated aqueous solution of brine (20 mL). The organic phase was dried (Na 2SO4) and solvent was removed under reduced pressure to give a syrup. Purification by chromatography (silica, 40 g) g with a gradient of 0-10% MTBE/heptanes gave the target compound (820 mg, 2.023 mmol, 89% yield) as a colorless oil. LCMS (Method r, Table 7) Rt 1.18 min, MS (DCI+) m/z 422.07, 424.09 [M+NH/]. 1H NMR (500 MHz, DMSO-d6) 8 9.94 (s, lH), 7.50 (dd, J 7.7, 1.6 Hz, lH), 7.42 - 7.36 (m, 2H), 7.36 -7.32 (m, 2H), 7.25 (t, J 7.8 Hz, lH), 7.17 (ddd, J 7.7, 1.7, 1.0 Hz, lH), 4.01 (s, 2H), 0.92 (s, 9H), 0.26 (s, 6H).
Step 6: sis of tert-butyl (3-(4-formylhydroxybenzyl)phenyl)carbamate NH2C02!Bu XPhos Gen 2 Precat.
Cs2C03, p-dioxane OH �Br 100 °C; then TBAF �NHBoc OHC� V OHC� V Nitrogen was sparged h a mixture of 4-(3-bromobenzyl)((tertbutyldimethylsily l)oxy)benzaldehyde (0.820 g, 2.023 mmol), tert-butyl ate (0.3027 g, 2.58 mmol), Cs2CO3 (1.006 g, 3.09 mmol) in p-dioxane (16 mL) for 30 min. Added the 2nd generation XPhos precatalyst (0.0937 g, 0.119 mmol) and continued sparging for 5 min, whereupon the reaction was heated to I 00 °C for 4 h. The reaction was cooled to room temperature, treated with a I N aqueous on of HCl (25 mL), and was extracted with MTBE (25 mL x 3). The combined organics were washed with brine (30 mL), dried over Na2SO4 and solvent was removed under d pressure. The residue was redissolved in THF (16 mL, 0.125 M) and d with TBAF/SiO2 (1.0-1.5 mmol/g, 4.1338 g, 4.13-6.2 mmol) for 45 min, whereupon solvent was removed under reduced pressure. Purification by chromatography (silica, 40 g) eluting with a gradient of 0-75% MTBE/heptanes gave tert-butyl (3-(4- formylhydroxybenzyl)phenyl)carbamate (380 mg, 1.161 mmol, 57% yield) was isolated as a gummy foam. LCMS (Method r, Table 7) Rt= 0.85 min; MS (DCI+) m/z = 345.0 [M+NH/]. 1H NMR (500 MHz, 6) 8 10.04 (s, IH), 9.86 (s, IH), 9.25 (s, IH), 7.37 (s, IH), 7.34 - 7.28 (m, 2H), 7.27 - 7.20 (m, 2H), 7.14 (t, J 7.8 Hz, IH), 6.82 (dt, J 7.7, 1.2 Hz, IH), 3.89 (s, 2H), 1.45 (s, 9H).
Step 7: Synthesis of (2S,6aS,6bR,7S,8aS,8bS,10R,l laR,12aS,12bS)-l 0-( 4-(3-aminobenzyl)- 3-hydroxyphenyl)-2,6b-difluo rohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- 6a, 6b, 7,8,8a,8b, I la,12, 12a,12b-decahydro-1H-naphtho[2', ]indeno[l,2-d][l,3]dioxol-4(2H)-one HO NH2 HO O HO OH TfOH ...?:JO ....OH NHBoe MgS04, ACN ,-70 + �I ""' I ""' ..... o OHC .& .& Triflic acid (0.060 mL, 0.680 mmol) was added drop-wise to a 0 °C slurry of (6S,8S,9R I0S, 1IS,13S,14S,16R l7S)-6,9-difluoro- l l,16, l7-trihydroxy- l7-(2-hydroxyacetyl)-10,13- dimethyl-6,7,8,9,10,11,12,13,14,15,16, l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (0.056 g, 0.136 mmol), utyl (3-(4-formylhydroxybenzyl)phenyl)carbamate (0.049 g, 0.150 mmol) and MgSO4 (0.049 g, 0.408 mmol) in MeCN (1.5 mL), maintaining a reaction temperature of <5 °C. After 30 min the reaction mixture was diluted with EtOAc (15 mL), and was washed sequentially with a saturated aqueous solution of NaHCO 3 (5 mL X 2), and then with a saturated aqueous solution of brine (3 mL).
The organic phase was dried (Na 2SO4) and solvent was removed under reduced pressure. Purification by e phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 30 mm column). A nt of MeCN (A) and 0.1% formic acid in water (B) was used, at a flow rate of 60 mL/min (0-5.0 min 15% A, .0-20.0 min linear nt 15-80% A, hold 5 min). Combined fractions were concentrated under reduced pressure to remove volatile solvents, and were then lyophilized to give the title compound as a white amorphous solid (6.7 mg). LCMS (Method r, Table 7) Rt = 0.70 min; MS m/z = 622.39 [M+H+]. 1H NMR (501 MHz, DMSO-d6) 8 9.51 (s, lH), 7.25 (d, J= 10.2 Hz, lH), 6.96 (d, J= 7.7 Hz, lH), 6.89 - 6.81 (m, 2H), 6.75 (d, J= 7.7 Hz, lH), 6.37 - 6.24 (m, 4H), 6.11 (s, lH), 5.63 (ddd, J= 49.2, 11.2, 6.4 Hz, lH), 5.50 (d, J 4.3 Hz, lH), 5.30 (s, lH), 5.07 (s, lH), 4.91 (d, J 4.8 Hz, lH), 4.85 (s, 2H), 4.47 (d, J 19.3 Hz, lH), 4.21 4.14 (m, 2H), 3.70 - 3.60 (m, 2H), 2.69 - 2.50 (m, lH), 2.26 (s, lH), 2.31 - 2.16 (m, lH), 2.07 - 1.94 (m, lH), 1.68 (q, J 10.2, 8.9 Hz, 2H), 1.64 1.50 (m, lH), 1.48 (s, 3H), 0.84 (s, 3H).
Example 8: Synthesis of (6aR,6bS,7S,8aS,8bS,1OR, l laR,12aS,12bS)- l0-(4-(3-aminobenzyl) hydroxyphenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl-6a,6b,7,8,8a,8b, l la,12, 12a,12bdecahydro-1H-naphtho [2', l ':4,5]indeno[ 1,2-d][ l,3]dioxol-4(2H)-one OH OH TfOH � .. ,,OH + NHBoc MgSO4, ACN Triflic acid (0.35 mLI, 3.83 mmol) was added drop-wise to a 0 °C slurry of (8S,9S,10R, l lS,13S,14S,16R,17S)-l l,16,17-trihydroxy(2-hydroxyacetyl)-10,13-dimethyl- 9,10, l l,12,13,14,15,16, ecahydro-3H-cyclopenta[a]phenanthrenone (0.296 g, 0.786 mmol), tert-butyl (3-(4-formyl-2 -hydroxybenzyl)phenyl)carbamate (0.251 g, 0.767 mmol) and MgSO4 (0.332 g, 2.76 mmol) in MeCN (3.0 mL), maintaining a reaction temperature of <5 °C. After 40 min the reaction was diluted with EtOAc (15 mL), and was washed sequentially with a saturated aqueous on of NaHCO 3 (10 mL X 2), and then with a saturated aqueous solution ofbrine (5 mL). The organic layer was dried over Na2SO4 and solvent was removed under reduced pressure. cation by chromatography (silica, 12 g) eluting with a gradient % MeOH/DCM gave the title compound (238.4 mg, 0.407 mmol, 53% yield) as a white solid. A portion ofthis material (ca. 79.1 mg) was further purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 X 30 mm column). A gradient of MeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of60 mL/min (0-5.0 min 15% A, 5.0-20 min linear gradient 15-60% A, hold 2 min). ed fractions were concentrated under reduced re to remove volatile solvents, and the resulting solution was frozen and lyophilized to give thetitle compound as an off-white solid (43.4 mg). LCMS (Method r, Table 7) R1 = 0.73 min; MS m/z = 586.2 [M+H+]. 1H NMR (501 MHz, DMSO-d6) 8 9.61 (s, lH), 7.30 (d, J 10.1 Hz, lH), 7.27 - 7.20 (m, lH), 7.04 (dd, J 7.7, 2.9 Hz, 2H), 6.95 - 6.91 (m, 2H), 6.90 (d, J 1.6 Hz, lH), 6.79 (dd, J 7.7, 1.6 Hz, lH), 6.15 (dd, J 10.1, 1.9 Hz, lH), 5.92 (d, J 1.6 Hz, lH), 5.29 (s, lH), 4.88 (d, J 5.1 Hz, lH), 4.79 (s, lH), 4.45 (d, J 19.4 Hz, lH), 4.28 (q, J 3.3 Hz, lH), 4.15 (d, J 19.4 Hz, lH), 3.82 (s, 2H), 2.59 - 2.49 (m, lH), 2.30 (dt, J= 13.0, 3.8 Hz, lH), 2.16 - 2.05 (m, lH), 2.07 - 1.98 (m, lH), 1.75 (d, J= 3.0 Hz, 2H), 1.73 -1.54 (m, 3H), 1.38 (s, 3H), 1.05 (qd, J 12.9, 4.8 Hz, IH), 0.97 (dd, J 11.2, 3.6 Hz, IH), 0.84 (s, 3H). e 9: Synthesis of (S)-N-(3-(4-((2S,6aS,6bR,7S,8aS,8bS,I OR, l laR, 12aS,12bS)-2,6b-Difluoro- 7- hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,Ila,12,12a,12b-dodecahydro- 1H-naphth o[2', l':4,5]indeno[1,2-d] [ l,3]dioxol- l0-yl)benzyl)phenyl)((S)(3-(2,5-dioxo-2,5-dihydro­ rol-l-yl)propanamido)propanamido)propanamide Step 1: Synthesis of (9H-Fluorenyl)methyl ((S)-l-(((S)((3-(4- ((2S,6aS,6bR,7S,8aS,8bS, I OR, l laR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyloxo-2 ,4,6a,6b,7,8,8a,8b,Ila, 12, b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d] [l,3]dioxol- l 0-yl)benzyl)phenyl)amino )oxopropanyl)amino)oxopropanyl)carbamate HO 2 0 �10 ,, Fmoc Ala AlaOH HATU, 2,6lul, THF 0"'" '""o � �,,i-o·� I O 0 FmocHN ,,,.. n 0 :::,... '° OH - � != 0 HATU (1.3 g, 3.41 mmol) and 2,6-lutidine (0.4 mL, 3.43 mmol) were added to a room temperature suspens10n of (2S,6aS,6bR, 7S,8aS,8bS, I OR, l laR, 12aS, 12bS)-l 0-(4-(3- aminobenzyl)phenyl)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- 6a,6b,7,8,8a,8b,Ila,12,12a,12b-decahydro-1H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one (1.0327 g, 1.705 mmol), and (S)((S)((((9H-fluoren yl)methoxy)carbonyl)amino)propanamido)propanoic acid (0.782 g, 2.046 mmol) in THF (11.5 mL). After 3 hours at room temperature, the reaction was diluted with EtOAc (16 mL), then washed sequentially with a IN aqueous solution of HCl (4 mL x 3), and a saturated aqueous solution of brine (4 mL). Purification by chromatography (silica, 40 g) eluting with a nt of 75-100% EtOAc/heptanes gave the title compound (0.926 g, 0.955 mmol, 56% yield). LC-MS d r, Table 7) Rt 1.01 min, m/z 970.18 [M+H+].1H NMR (500 MHz, DMSO-d6) 8 9.85 (d, J 5.6 Hz, IH), 8.08 (d, J 7.3 Hz, IH), 7.89 (dd, J 7.5, 1.0 Hz, 2H), 7.76 -7.69 (m, 2H), 7.55 (d, J 7.4 Hz, IH), 7.49 - 7.16 (m, 13H), 6.94 - 6.88 (m, IH), 6.30 (ddd, J IO.I, 3.7, 1.9 Hz, IH), 6.14 (dt, J 2.6, 1.2 Hz, IH), 5.74 - 5.55 (m, IH), 5.53 (dt, J .0, 2.5 Hz, IH), 5.12 (t, J= 6.0 Hz, IH), 4.95 (d, J= 5.1 Hz, IH), 4.52 (dd, J= 19.4, 6.2 Hz, IH), 4.38 (p, J= 7.0 Hz, IH), 4.32 -4.16 (m, 5H), 4.09 (p, J= 6.9 Hz, IH), 3.88 (d, J= 10.9 Hz, 2H), 2.65 - 2.60 (m, IH), 2.33 -2.20 (m, IH), 2.05 (d, J 13.5 Hz, IH), 1.77 -1.63 (m, 3H), 1.50 (s, 3H), 1.28 (d, J 7.l Hz, 3H), 1.23 (d, J 7.l Hz, 4H), 0.88 (d, J 12.6 Hz, 3H).
Step 2: Synthesis of(S)amino-N-((S)((3-(4- ((2S,6aS,6bR7S,8aS,8bS, 1 OR, l laR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyloxo-2 ,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d] [l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)propanamide �JOJ),, '' ' O" H2N� � � OH 0 - Diethylamine (0.5 mL, 4.79 mmol) was added to a room ature solution of(9H-fluoren- 9-yl)methyl ((S)- l-(((S)- (4-((2S,6aS,6bR7S,8aS,8bS,l0R,l laR12aS,12bS)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro- hth o[2', l':4,5]indeno[l,2-d] [l,3]dioxol- benzyl)phenyl)amino)oxopropanyl)amino) oxopropanyl)carbamate (1.18 g, 1.216 mmol) in THF (6.0 mL). After 2 h, MTBE (10 mL) was added, which resulted in the immediate precipitation of a yellow solid. This slurry was stirred for 90 min, filtered, and washed with MTBE (5 mL x 3) to give a yellow solid (802.7 mg). This material was purified further by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 50 mm column). A gradient ofMeCN (A) and 0.1 % formic acid in water (B) was used, at a flow rate of90 mL/min (0-5.0 min 15% A, .0-20.0 min linear gradient 15-75% A, hold 2 min, 22.0-22.5 min linear gradient from 75-95%, hold for 4 min). Combined fractions were concentrated under d pressure to dryness and then dried overnight in the vacuum oven at 50 °C. LC-MS (Method r, Table 7) R1 = 0.76 min, m/z = 748.5 [M+H+]. 1H NMR indicates that the title compound 1s an approximately 1: 1 mixture with (2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l0-(4-(3-((S)((S)aminopropanamido) propanamido)benzyl)phenyl)-2,6b-difluo rohydroxy-6a,8a-dimethyloxo- 2, 4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxole-8bcarboxylic acid. (0.170 g total, 0.115 mol and 10% yield of each nd). 1H NMR (500 MHz, DMSO-d6) 8 10.00 (d, J= 3.3 Hz, 2H), 8.42 (d, J= 34.2 Hz, 2H), 8.30 (s, lH), 7.48 7.37 (m, 3H), 7.38 7.29 (m, 5H), 7.31 7.15 (m, 8H), 6.92 (d, J= 7.6 Hz, 2H), 6.28 (ddd, J= 10.3, 6.6, 1.9 Hz, 3H), 6.12 (d, J= 3.7 Hz, 3H), 5.77 5.53 (m, 3H), 5.45 (d, J= 7.7 Hz, 3H), 5.04 4.99 (m, lH), 4.94 (d, J= 5.1 Hz, lH), 4.50 (d, J= 19.4 Hz, lH), 4.40 (s, 3H), 4.23 4.12 (m, 2H), 3.54 (dq, J= 17.6, 6.9 Hz, lH), 2.71 2.56 (m, lH), 2.30 2.15 (m, lH), 2.03 (d, J= 14.2 Hz, 2H), 1.94 (d, J= 14.3 Hz, lH), 1.84 (d, J= 14.1 Hz, lH), 1.76 1.59 (m, 7H), 1.49 (d, J= 2.6 Hz, 8H), 1.39 1.10 (m, 13H), 1.00 (s, 4H), 0.86 (s, 3H).
Step 3: Synthesis of (3-(4-((2S,6aS,6bR,7S,8aS,8bS,1 OR,llaR,12aS,12bS)-2,6b- Difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl oxo-2,4,6a,6b,7,8,8a,8b,l la,12, 12a, ecahydro-1H-naphtho [2', l ':4,5]indeno[1,2-d][ l,3]dioxol- l0-yl)benzyl)phenyl)((S)(3-(2,5-dioxo- 2,5-dihydro-lH-pyrrol- l-yl)propanamido )propanamido)propanamide ��O-N� 0 O µ i-Pr2NEt, DMF Diisopropylethyl amine (0.1 mL, 0.573 mmol) was added to a room temperature solution of (S)amino-N-((S)((3-(4-((2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)-2,6b-difluoro- 7-hydroxy- 8b-(2-hydroxyacetyl)-6a,8a -dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H­ naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)propanamide (0.170 g, 0.227 mmol) and N-succinimidyl 3-maleimidopropionate (0.0691 g, 0.260 mmol) in DMF (2.5 mL). After 30 min, the pH ofthe reaction mixture was adjusted to 4-5 by drop-wise on ofa 7% solution ofTFA in water (1.2 mL). The crude mixture was purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 50 mm column). A gradient ofMeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of90 mL/min (0-5.0 min 15% A, 5.0-20 min linear gradient 15-85% A, hold 2 min). Combined fractions were concentrated under reduced pressure to remove volatile solvents, and the resulting solution was fro zen and lyophilized to give a white solid (85.2 mg, 0.0473 mmol, 21 % yield). LC-MS (Method R Table 7) R1 = 0.82 min, m/z = 899.92 [M+H+]. 1H NMR data was consistent with a 1: 1 mixture ofthe title compound S,6bR, 7S,8aS,8bS,10RllaR12aS,12bS)(4- (3-((S )( (S)(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol- lyl )propanamido)propanamido)propanamido)benzyl)phenyl)-2,6b-difluo rohydroxy-6a,8a-dimethyl oxo- l ,2, 4,6a,6b,7,8,8a, l la,12, 12a,12b-dodecahydro-8bH-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxole-8bcarboxylic acid (see e 10 for an alternative preparation ofthe title compound, which avoids this mixture). . MS analysis confirms that this material is a mixture oftwo compounds with m/z = 899.1 [M+H+] and m/z = 885.0 [M+H+]. 1H NMR (400 MHz, DMSO-d 6) 8 9.71 (s, 2H), 8.16 (d, J = 7.1 Hz, 2H), 8.03 (d, J= 7.3 Hz, 2H), 7.49 7.29 (m, 9H), 7.30 7.13 (m, 9H), 6.96 (s, 3H), 6.92 6.85 (m, 2H), 6.27 (dt, J 10.1, 1.9 Hz, 2H), 6.11 (d, J 2.3 Hz, 2H), 5.74 5.53 (m, 2H), 5.46 (d, J 23.9 Hz, 4H), 4.93 (d, J 5.0 Hz, lH), 4.32 (p, J 7.1 Hz, 2H), 4.27 4.13 (m, 3H), 4.17 (s, 3H), 3.59 (t, J 7.3 Hz, 4H), 2.69 2.53 (m, 2H), 2.38 (t, J= 7.3 Hz, 4H), 2.28 (s, 3H), 2.22 (s, lH), 2.08 1.98 (m, lH), 1.98 1.90 (m, lH), 1.83 1.68 (m, 2H), 1.69 (s, 2H), 1.66 (s, 2H), 1.48 (d, J= 3.7 Hz, 8H), 1.25 (d, J= 7.0 Hz, 6H), 1.15 (d, J= 7.1 Hz, 6H), 0.99 (s, 3H), 0.84 (s, 3H).
Example 10: Synth esis of (S)-N-(3-(4-((2S,6aS,6bR7S,8aS,8bS,l0R, l laR,12aS,12bS)-2,6b-Difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7 ,8,8a,8b, Ila, 12, 12a,12b-dodecahydro- 1H-naphth o[2', l ':4,5]indeno[1,2-d][ l,3]dioxol- l 0-yl)benzyl)phenyl)((S)(3-(2,5-dioxo-2,5-dihydro­ IH-pyrrol- l -yl)propanamido )propanamido)propanamide Step 1: Synthesis of tert-butyl ((S)(((S)((3-(4-((2S,6aS,6bR7S, 8aS,8bS, I OR l laR 12aS, 12bS)-2,6b-difluorohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dime -oxo- 2,4,6a,6b,7,8,8a,8b, Ila,12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol- l 0- yl)benzyl)phenyl)amino)oxopropanyl)amino)oxopropanyl)carbamate : O Boe Ala Ala OH � HATU, 2,6 lut, THF ' � r(¾i ,,,LO lH O � � BocHN ,.. N n 0� :::,__ OH HATU (610 mg, 1.605 mmol) and 2,6-lutidine (0.3 mL, 2.58 mmol) were added to a room temperature mixture of (2S,6aS,6bR,7S,8aS,8bS,I OR l laR,12aS,12bS)-l 0-(4-(3-aminobenzyl)phenyl)- 2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl-6a,6b,7,8,8a,8b,Ila, 12,12a,12bdecahydro-1H-naphtho [2', l indeno[l,2-d][l,3]dioxol-4(2H)-one (648.1 mg, 1.070 mmol), and (S) ((S)((tert-butoxycarbonyl)amino)propanamido)propanoic acid (334 mg, 1.284 mmol) in THF (11.5 mL). After 9 hours the reaction was diluted with EtOAc (16 mL), then washed with a IN aqueous solution of HCl (4 mL x 3), followed by a saturated aqueous solution of brine (4 mL). Purification by chromatography (silica, 40 g) eluting with a gradient of 0-10% MeOH/DCM gave the title nd as a yellow foam (773.7 mg, 0.912 mmol, 85% yield). LC-MS d r, Table 7) R1 = 0.92 min, m/z = 848.53 [M+H+]. 3] Step 2: Synthesis of amino-N-((S)((3-(4- ((2S,6aS,6bR7S,8aS,8bS, IOR,l laR, 12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyl oxo-2,4,6a,6b,7,8,8a,8b, Ila, 12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2- d][l ,3]dioxol- l 0-yl)benzyl)phenyl)amino)oxopropanyl)propanamide l.o"" i,O___,O� OS 0 : o' TFA (1.97 mL, 25.6 mmol) was added drop-wise to a room temperature solution of tert-butyl ((S)- l-(((S)((3-(4-((2S,6aS,6bR,7S,8aS,8bS, I OR l laR,12aS,12bS)-2,6b-difluorohydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, Ila,12,12a,12b-dodecahydro-1H- naphtho[2', l':4,5]indeno[ [ l,3]dioxol-l 0-yl)benzyl)phenyl)amino )oxopropanyl)amino) oxopropanyl)carbamate (0.7683 g, 0.906 mmol) in DCM (6.0 mL). After 50 min solvent was removed under reduced pressure to give a brown syrup. The residue was dissolved in 1:I DMSO:MeOH (12 mL) and purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 50 mm column).
A gradient ofMeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of90 mL/min (0-5.0 min % A, 5.0-20 min linear gradient 15-75% A, hold 2 min, 22.0-22.5 min linear gradient 75-95% A, hold 4 min). Combined fractions were concentrated under reduced pressure to dryness and the residue was dried overnight in the vacuum oven at 50 °C to give the title compound (230 mg, 0.308 mmol, 34% yield. LCMS (Method r, Table 7) major acetal isomer Rt 0.73 min, m/z 748.78 . 1H NMR (400 MHz, DMSO-d6) 8 10.01 (s, IH), 8.62 (d, J 7.2 Hz, IH), 8.04 (d, J 5.4 Hz, 3H), 7.46 -7.31 (m, 4H), 7.31 - 7.13 (m, 4H), 6.91 (d, J 7.6 Hz, IH), 6.27 (dd, J 10.2, 1.9 Hz, IH), 6.11 (s, IH), 5.76 - 5.47 (m, 2H), .43 (s, IH), 4.93 (d, J 4.6 Hz, IH), 4.49 (d, J 19.5 Hz, IH), 4.42 (q, J 7.l Hz, IH), 4.23 - 4.13 (m, 2H), 2.72 - 2.54 (m, IH), 2.33 - 2.16 (m, 2H), 2.02 (dt, J 13.6, 3.6 Hz, IH), 1.69 (h, J 5.9, 5.1 Hz, 3H), 1.48 (s, 4H), 1.33 (d, J 7.0 Hz, 3H), 1.30 (d, J 7.l Hz, 3H), 0.85 (s, 3H).
Step 3: Synthesis of (S)-N-(3-(4-((2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)-2,6b­ Difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,Ila,12,12a,12bdodecahydro-1H-naphtho [2', I':4,5]indeno[1,2-d][ l,3]dioxol- l 0-yl)benzyl)phenyl)((S)(3-(2,5-dioxo- 2,5-dihydro-IH-pyrrol-l-yl)propanamido)propanamido)propanamide ,-::; ��O_NJ( o"' 0 O µ l,,OJ) '--'--o 0 i-Pr2NEt, DMF Diisopropylethylamine (0.1 mL, 0.573 mmol) was added to a room ature on of (S)amino-N-((S)- (4-( (2S,6aS,6bR,7S,8aS,8bS, IOR, l aS,12bS)-2,6b-difluoro- 7-hydroxy- 8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H­ naphtho[2', l ':4,5]indeno[1,2-d][ l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)propanamide (0.220 g, 0.294 mmol) and N-succinimidyl 3-maleimidopropionate (0.086 g, 0.324 mmol) in DMF (2.8 mL). After 30 min the pH ofthe reaction mixture was adjusted to 4-5 by drop-wise addition of a 7% solution of TFA in water (1.0 mL). The crude mixture was purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 50 mm ). A gradient ofMeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of90 mL/min (0-5.0 min 15% A, 5.0-20 min linear gradient 15-85% A, hold 2 min). Combined fractions were concentrated under reduced pressure to remove volatile solvents, and the resulting solution was frozen and lyophilized to give a white solid (175.2 mg, 0.195 mmol, 66% yield). LC-MS (Method r, Table 7) Rt 0.82 min, m/z 899.87 [M+H+]. 1HNMR (400 MHz, DMSO-d6) 8 9.70 (s, IH), 8.14 (d, J= 7.0 Hz, IH), 8.01 (d, J= 7.2 Hz, IH), 7.47- 7.35 (m, 2H), 7.32 (d, J= 8.1 Hz, 2H), 7.26 - 7.10 (m, 4H), 6.95 (s, IH), 6.87 (dt, J= 7.6, 1.3 Hz, IH), 6.26 (dd, J= 10.2, 1.9 Hz, IH), 6.09 (d,J= 2.0 Hz, IH), 5.72- 5.51 (m, IH), 5.48 (s, IH), 5.41 (s, IH), 4.91 (d,J= 4.9 Hz, IH), 4.47 (d, J= 19.4 Hz, IH), 4.30 (p, J= 7.1 Hz, IH), 4.25- 4.11 (m, 3H), 3.85 (s, 2H), 3.57 (t, J= 7.3 Hz, 2H), 2.71- 2.48 (m, IH), 2.36 (dd, J= 8.0, 6.7 Hz, 2H), 2.23 (ddt, J= 25.l, 12.2, 6.6 Hz, 2H), 2.01 (dt, J = 13.7, 3.7 Hz, IH), 1.75- 1.57 (m, 3H), 1.48 (p, J= 11.9 Hz, IH), 1.46 (s, 3H), 1.24 (d, J= 7.2 Hz, 3H), 1.13 (d,J= 7.2 Hz, 3H), 0.83 (s, 3H).
Example 11: Synthesis of ((S)-l-(((S)-l-((3-(4-((2S,6aS,6bR7S,8aS,8bS,l0R,l laR,12aS,12bS)- 2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,Ila,12,12a, 12bdodecahydro-1H-naphtho [2',l':4,5]indeno[l,2-d][l,3]dioxol- l 0-yl)benzyl)phenyl)amino)oxopropan yl)amino)-l-oxopropanyl)(3-(2,5-dioxo-2,5-dihydro-IH-pyrrol- l-yl)propanamido)(IH-imidazol- 4-yl)propanamide Stepl: sis of (S)((tert-Butoxycarbonyl)amino)(IH-imidazolyl)propanoic acid, 2 hydrochloric acid HN-.§ To a solution of (S)amino(IH-imidazolyl)propanoic acid (1.55 g, 9.99 mmol) in water (40 mL) and 1,4-dioxane (10 mL) at 0°C were added NaOH (10 mL, 19.98 mmol) and BOC­ anhydride (2.319 mL, 9.99 mmol). The resulting mixture was stirred at 23°C for 4h. Then the mixture was acidified with HCl solution to pH 5, and washed with EtOAc (3 X 30 mL). Then the inorganic layer was freeze-dried to give the title compound (including NaCl)) (4.449 g, 9.90 mmol, 99 % yield) as a white solid. LCMS (Method m, Table 7) R1 1.22 min, m/z 256.2 (M+1t. 9] Step 2: Synthesis of tert-butyl -(((S)-l-(((S)-l-((3-(4-((2S,6aS,6bR7S, 8aS,8bS,1 OR l laR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol- l 0- yl)benzyl)phenyl)amino)oxopropanyl)amino)oxopropanyl)amino)(IH-imidazolyl)-loxopropanyl )carbamate oo �E o Xt � NJ H O lj C � A' YH )-N� p- ' . � 0 f HN� o••· 1"' .N - IL.) �"•-.Lo• OH HN...§ To a solution of ((tert-butoxycarbonyl)amino)(IH-imidazolyl)propanoic acid, 2hydrochloric acid (170 mg, 0.197 mmol), (S)((tert-butoxycarbonyl)amino)(IH-imidazol yl)propanoic acid, 2 hydrochloric acid (443 mg, 0.986 mmol) in THF (20 mL) at 0°C were added DIPEA (0.345 mL, 1.973 mmol) and HATU (90 mg, 0.237 mmol), DMAP (31.3 mg, 0.256 mmol) and the resulting e was stirred at 0°C for 10 min, and gradually warmed to 25°C for 16h. After that, the mixture was concentrated to give the e, which was purified by DCM/MeOH(l0:l) by silica gel to obtain the title compound (194 mg, 0.138 mmol, 69.9 % yield) as a yellow solid. LCMS (Method m, Table 7) R1 1H NMR: (400 MHz, DMSO-d6) 8 ppm: 0.82-0.89 (m, 1.72 min, m/z 985.3 (M+l)\ l0H), 1.12-1.18 (m, 9H), 1.23 (s, 9H), 1.68-1.71 (m, 2H), 2.20-2.33 (m, 2H), 3.86-3.88 (m, 2H), 4.18-4.29 (m, 4H), 4.36-4.39 (m, lH), 4.49-4.54 (m, lH), 4.94 (d, J= 4.4 Hz, lH), 5.13 (bs, lH), 5.45 (s, lH), 5.57-5.74 (m, 2H), 6.12(s, lH), 6.29 (d, J= 10.0 Hz, lH), 6.91 (d, J= 8.0 Hz, lH), 7.01 (d, J= 8.0 Hz, lH), 7.10-7.25 (m, 4H), 7.29-7.37 (m, 3H), 7.44-7.49 (m, 2H), 8.06 (d, J= 6.0 Hz, lH).
Step 3: Synthesis of (S)amino-N-((S)- l-(((S)- l-((3-(4-((2S,6aS,6bR7S,8aS,8bS, 1 OR l laR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b, l la, ,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l 0- yl)benzyl)phenyl)amino)oxopropanyl)amino)oxopropanyl)( lH-imidazolyl)propanamide, uoroacetic acid oo E o XiJ � _J H O N � C rP' B E o YH )-N---,.J( ,,., ' H 0 <P' � 0 f HN�V 0'"� H F-+-{ � �"··Lo• OH 0 H2N✓N��- ,? E B Iv.N F HN-# ',-'\ V /?' o �O fHN� 0'"� - �"··Lo• OH OH HN...# 2] To a solution of tert-butyl ((S)(((S)(((S)( (3-(4- ((2S,6aS,6bR7S,8aS,8bS, 1 OR, l laR, 12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyloxo-2 ,4,6a,6b,7,8,8a,8b, l la, 12, 12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d][l,3]dioxol- l 0-yl)benzyl)phenyl)amino opropanyl)amino)oxopropanyl)amino)( lH­ imidazolyl)- l-oxopropanyl)carbamate (120 mg, 0.122 mmol) in DCM (3 mL) was added TFA (0.6 mL, 7.79 mmol), and the reaction mixture was stirred at 20°C for 1 hour. After that, the mixture was diluted with DCM, concentrated in vacuo at about 25°C to give the title compound (149 mg, 0.103 mmol, 84.90 % yield) as a yellow solid. LCMS (Method m, Table 7) R1 1.64 min, m/z 885.3 (M+It.
Step 4: sis of (S)-N-((S)- l-(((S)((3-(4- ((2S,6aS,6bR7S,8aS,8bS,1 OR, l laR,12aS,12bS)-2,6b-difluorohydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyloxo-2 ,4,6a,6b,7,8,8a,8b, l la, 12, 12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d][l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)amino)- l-oxopropanyl)(3-(2,5-dioxo- 2,5-dihydro- rol- l-yl)propanamido)( lH-imidazolyl)propanamid DIPEA (0.106 mL, 0.607 mmol) was added to the solution of (S)amino-N-((S)-l-(((S) -((2S,6aS,6bR7S,8aS,8bS,IOR, l laR,12aS,12bS)-2,6b-difluoro- oxy-8b-(2-hydroxyacetyl)- 6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,Ila, 12, 12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2- d][l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)amino)oxopropanyl)(IH-imidazol- -yl)propanamide, 3trifluoroacetic acid (149 mg, 0.121 mmol) and 2,5-dioxopyrrolidin-l-yl 3-(2,5-dioxo- 2,5-dihydro-IH-pyrrol- l-yl)propanoate (48.5 mg, 0.182 mmol) in DMF (2 mL) at 0°C, and then the mixture was stirred at room temperature for 2 h. The on e was purified by prep-HPLC (Mobile Phase:A=0.05%TFA in water, B=MeCN; Flow Rate: 2mL/min) to afford the title compound (11.4 mg, 9.02 mmol, 7.43 % yield) as a white solid. LCMS (Method m, Table 7) RT= 1.62 min, m/z 1058.3 (M+Na)\ 1H NMR: (400 MHz, ) 8 ppm: 0.86 (s, 3H), .29 (m, 9H), 1.46-1.54 (m, 3H), .76 (m, 2H), 1.98-2.06 (m, 2H), 2.20-2.33 (m, 2H), 2.40-2.44 (m, 2H), 2.60-2.68 (m, IH), 2.88-2.94 (m, IH), 3.00-3.05 (m, IH), 3.57 (t, J 7.4 Hz, 2H), 3.89 (s, 2H), 4.18-4.39 (m, 4H), 4.49-4.60 (m, 2H), 4.95 (d, J 4.8 Hz, IH), 5.46 (s, IH), .74 (m, 2H), 6.13 (s, IH), 6.30 (d, J 10.4 Hz, IH), 6.92 (d, J 7.6 Hz, IH), 7.00 (s, 2H), 7.20-7.28 (m, 4H), 7.35-7.46 (m, 5H), 8.16 (d, J 6.4 Hz, IH), 8.23 (d, J 7.2 Hz, IH), 8.31 (d, J 8.4 Hz, IH), 8.96 (s, IH), 9.89 (s, IH).
Example 12: Synthesis of (S)(((S)- l-((4-((4-((2S,6aS,6bR7S,8aS,8bS,l0R,1laR,12aS,12bS)-2,6b­ difluorohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b, 7,8,8a,8b,Ila, 12, 12a, 12bdodecahydro-1H-naphtho [2', l':4,5]indeno[l,2-d][l,3]dioxol- l 0-yl)phenyl)thio)phenyl)amino) oxopropanyl)amino)(3-(2,5-dioxo-2,5-dihydro-IH-pyrrol-l-yl)propanamido)oxopentanoic acid (Cpd. No. 81) Step 1: Synthesis of (2S,6aS,6bR7S,8aS,8bS,I OR l laR12aS,12bS)- l 0-( 4-( ( 4- Aminophenyl)thio )phenyl)-8b-(2-((tert-butyldimethyl silyl)oxy)acety1)-2,6b-difluorohy droxy-6a,8adimethyl-6a ,6b,7,8,8a,8b,Ila, 12, 12a,12b-decahydro-1H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxol- 4(2H)-one OH XS/O' ....07·",,,o hQ l,,, / N 0- ....07"•.,,o h 0 NH2 Q F 0 NH2 To a stirred solution of (2S,6aS,6bR,7S,8aS,8bS,1 OR l laR 12aS,12bS)-l 0-( 4-(( 4- aminophenyl)thio)phenyl)-2, 6b-difluorohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- 6a, 6b,7,8,8a,8b,Ila, 12, 12a,12b-decahydro-1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one (62.4 mg, 0.1 mmol) and imidazole (34.0 mg, 0.500 mmol) in DCM was added TBS-Cl (45.2 mg, 0.300 mmol) at 0°C, After stirring was continued for 30 min at the same temperature, the mixture was allowed to warm to room temperature and stirred for 2 h. the reaction mixture was diluted with EtOAc(l0 mL), washed with water (2xl0 mL) and brine(lxl0 mL), dried over Na2S04, fil tered and concentrated in vacuo.
The residue was purified by column (EA:PE= 1:10-1:1) to give the product (2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l 0-(4-((4-aminophenyl)thio ) phenyl)-8b-(2-((tertbutyl dimethylsilyl)oxy)acetyl)-2,6b-difluoro- 7-hydroxy-6a,8a-dimethyl-6a,6b,7,8,8a,8b, lla,12, 12a, 12bdecahydro-1H-naphtho [2',l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one (50 mg, 0.068 mmol, 67.8 % yield).
LCMS (Method m, Table 7) R1 2.144 min, m/z 738 (M+H).
Step 2: sis of utyl ((S)((4-((4- ( (2S,6aS,6bR7S,8aS,8bS,1 OR, l laR,12aS,12bS)-8b-(2-((tert-butyl dimethyl silyl)oxy)acetyl)-2,6bdifluoro- 7-hydroxy-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H­ naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l henyl)thio)phenyl)amino)oxopropanyl)carbamate )(. ,, ....07·"0-h.,,o Q 0 NH2 CI-P � F 0 \: N 0 To a stirred solution of S,6bR,7S,8aS,8bS,1OR llaR12aS,12bS)- l 0-( 4-(( 4- aminophenyl)thio)phenyl)-8b-(2-( (tert-butyldimethylsilyl)oxy )acetyl)-2, 6b-difluorohydroxy -6a,8adimethyl-6a ,6b,7,8,8a,8b,l la,12, 12a,12b-decahydro-1H-naphtho[2', l ':4,5] [1,2-d][ l ,3]dioxol- 4(2H)-one (0.148 g, 0.2 mmol) and (S)((tert-butoxycarbonyl) amino)propanoic acid (0.076 g, 0.400 mmol) in DCM (3 mL) was added pyridine (0.162 mL, 2.000 mmol), fol lowed by POCh (0.075 mL, 0.800 mmol) in dropwise. The reaction mixture was stirred for 1 hour at ambient temperature, then concentrated in vacuo, and the residue was purified by column (EA:PE=1: 10-9:1) to give tert-butyl ((S) (( ((2S,6aS,6bR,7S,8aS,8bS,1OR,llaR12aS,12bS)-8b-(2-((tert-butyl dimethylsilyl) oxy)acetyl)-2,6bdifluoro- 7-hydroxy-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-1H­ naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l 0-yl)phenyl)thio)phenyl)amino)oxopropanyl)carbamate (0.073 g, 0.080 mmol, 40 % yield) as a olid. LCMS (Method m, Table 7) R1 2.156 min, m/z 909 (M+H).
Step 3: Synthesis of (S)amino-N-(4-((4-((2S,6aS,6bR,7S,8aS,8bS,1OR l laR,12aS,12bS)- 2,6b-difluorohydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la,12,12a,12bdodecahydro-1H-naphtho [2', l ':4,5]indeno[1,2-d][ 1,3]dioxolyl)phenyl)thio)phenyl)propanamide To a stirred solution of tert-butyl ((S)- l-((4-((4-((2S,6aS,6bR7S,8aS,8bS,l0R,l laR l2aS,l2bS)-8b-(2-((tert-butyldimethylsilyl)oxy)acetyl)-2,6b-difluoro hydroxy-6a,8a-dimethyloxo- 2, 4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5] indeno[l,2-d] [l,3]dioxol- l 0- yl)phenyl)thio)phenyl)amino)oxopropanyl)carbamate (0.091 g, 0.1 mmol) in methylene de (1 mL) was added TFA (1 mL, 12.98 mmol), and the solution was stirred for 2 hours at ambient temperature, then concentrated m vacuo to give the product (S)amino-N-(4-((4- ((2S,6aS,6bR7S,8aS,8bS,1OR,l laR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyl oxo-2,4,6a,6b,7,8,8a, 8b, l la,12,12a,12b-dodecahydro-1H-naphtho[ 2', ]indeno[l,2- d][l,3]dioxol- l 0-yl)phenyl)thio)phenyl)propanamide (7.21 g, 10.38 mmol, 80 % yield). LCMS (Method m, Table 7) Rt 1.653 min, m/z 695 (M+H).
Step 4: Synthesis of (S)(tert-Butoxy)(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol- lyl )propanamido )oxopentanoic acid To a stirred solution of (S)amino(tert-butoxy)oxopentanoic acid (406 mg, 2 mmol) and oxopyrrolidin- l-yl 3-(2,5-dioxo-2,5-dihydro-lH-pyrrol- l-yl)propanoate (532 mg, 2.000 mmol) in yl formamide (2 mL) was added DIPEA (0.524 mL, 3.00 mmol). After ng was continued for 2h at room temperature, the reaction mixture was diluted with EtOAc (10 mL), washed with water (2 X 10 mL) and brine (lxlO mL), dried over Na2SO4, filtered and concentrated in vacuo, and the residue was purified by column (MeOH/DCM 0:10-1:10) to give the title compound (209 mg, 0.590 mmol, 29.5 % yield) as a yellow oil. LCMS d m, Table 7) Rt 1.490 min, m/z 377 (M+Na).
Step 5: Synthesis of (S)-tert-butyl 5-(((S)((4-(( 4-((2S,6aS,6bR7S,8aS, 8bS,1 OR llaR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b,l la,12,12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l 0- yl)phenyl)thio )phenyl)amino)oxopropanyl)amino)(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l - panamido )oxopentanoate :� .. ,Q-s,,o h �iNJN� (X}' N- .,)___ O\\ 0 o 0 N"­ � OH --¾( / 0 0 # F. � � H2N O A solution of (S)amino-N-(4-((4-((2S,6aS,6bR7S,8aS,8bS,1OR, l S,12bS)-2,6bdifluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12,12a,12bdodecahydro-1H-naphtho [2', l':4,5]indeno[l,2-d][l,3]dioxol-l0-yl)phenyl) thio)phenyl)propanamide ( 40 mg, 0.058 mmol), (tert-butoxy)(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol- l-yl)propanamido) oxopentanoic acid (30.6 mg, 0.086 mmol), HATU (32.8 mg, 0.086 mmol) and DIPEA (0.030 mL, 0.173 mmol) in dimethyl formamide ( 2 mL) was stirred overnight at room temperature, and diluted with EtOAc (10 mL), washed with water (2 Xl0 mL) and brine (lxlO mL), dried over Na2SO4, filtered and evaporated in reduced pressure. The residue was purified by column tography (MeOH/ DCM=0:10;1:10) to give the title nd (30 mg, 0.029 mmol, 50.5 % yield). LCMS (Method m, Table 7) R1 2.051 min, m/z 1031 (M+H).
Step 6: Synthesis of (S)(((S)- l-((4-((4-((2S,6aS,6bR,7S,8aS,8bS,l0R,l laR,12aS,12bS)- 2, 6b-Difluorohydroxy-8b-(2-hydroxyacetyl )-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,11a,12, 12a, l2bdodecahydro-1H-naphtho [2', l':4,5]indeno[l,2-d][l,3]dioxolyl)phenyl)thio)phenyl)amino) oxopropanyl)amino)(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol- l-yl)propanamido)oxopentanoic acid OH OH 0 F 0 To a stirred solution of (S)-tert-butyl 5-(((S)- l-( ( 4-(( 4-((2S,6aS,6bR,7S,8aS,8bS,1OR 11aR, l2aS, l 2bS )-2, 6b-difluorohydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-l 0- yl)phenyl)thio )phenyl)amino)oxopropanyl)amino)(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l - yl)propanamido)oxopentanoate (10.31 mg, 0.01 mmol) in DCM (0.5 mL) was added TFA (0.5 mL, 6.49 mmol). After stirring was ued for 2 h, the reaction mixture was concentrated in vacuo to give the title compound (6.83 mg, 7.00 µmol, 70 % yield). LCMS d m, Table 7) Rt 1.875 min, m/z 975 (M+H).
Example 13: Synthesis ofN-(4-(4-((2S,6aS,6bR,7S,8aS,8bS,l0R, l laR,12aS,12bS)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7 8b, l la,12,12a,12b-dodecahydro- 1H-naphth o[2', l indeno[l,2-d] [l,3]dioxol-l0-yl)phenoxy)phenyl)(3-(2,5-dioxo-2,5-dihydro-lH­ pyrrol- l-yl)propanamido)-3,6,9,12,15,18,21,24,27,30,33,36-dodecaoxanonatriacontanamide 7] Step 1: Synthesis of (2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)(4-( 4- henoxy)phenyl)-8b-(2-((tert-butyldimethy lsilyl)oxy)acety1)-2,6b-difluorohydroxy -6a,8adimethyl-6a ,6b, 7,8,8a,8b, l la,12,12a,12b-decahydro-1H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxol- 4(2H)-one [,,,, / To a stirred solution of (2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l 0-( 4-(4- aminophenoxy)pheny1)-2, 6b-difluorohydroxy -8b-(2-hydroxyacetyl)-6a,8a-dimethyl- 6a,6b, ,8b, l la,12,12a,12b-decahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][1,3] dioxol-4(2H)-one (290 mg, 0.477 mmol) and imidazole (162 mg, 2.386 mmol) in CH2Ch (10 mL) was added TBS-Cl (216 mg, 1.432 mmol) at 0°C, After stirring was continued for 30 min at the same temperature, the mixture was allowed to warm to room temperature and stirred for 2 h. the reaction mixture was diluted with EtOAc(10 mL), washed with water (2xl0 mL) and brine(lxl0 mL), dried over Na2S04, filtered and concentrated in vacuo. The residue was ed by column (EA:PE 1:10-9:1) to give title compound (300 mg, 0.416 mmol, 87 % yield). LCMS (Method m, Table 7) Rt 1.812 min, m/z 722 (M+H).
Step 2: Synthesis of tert-butyl (39-((4-(4-((2S,6aS,6bR, 7S,8aS,8bS, llaR,12aS,12bS)-8b-(2- ((tert-Butyldimethylsilyl)oxy)acetyl)-2,6b-difluoro- 7-hydroxy-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-l 0- yl)phenoxy)phenyl)amino)oxo-3,6,9,12, 15,18,21,24,27,30,33,36-dodecaoxanonatriacontyl)carbamate E 0 (' 0....._...,...._ o� o.-__)l oH o....._...,...._ o....._...,...._ o 0� 0� ) HN�o....._...,...._ 0....._...,...._ o� o C[-P�� 0 To a stirred solution of (2S,6aS,6bR7S,8aS,8bS,1 laR,12aS,12bS)(4-( 4- aminophenoxy)phenyl)-8b-(2-( (tert-butyl dimethylsilyl)oxy)acetyl)-2, 6b-difluorohydroxy-6a,8adimethyl-6a ,6b,7,8,8a,8b, l la, 12, 12a,12b-decahydro-1H-naphtho[2', l':4,5] indeno[ 1,2-d][ l,3]dioxol- one (144 mg, 0.2 mmol) and 2,2-dimethyloxo-3,8, l l,14,17,20,23,26,29,32,35,38,41-tridecaoxa- -azatetratetracontanoic acid (144 mg, 0.200 mmol) in CH2Ch (3 mL) was added pyridine (0.162 mL, 2.000 mmol), ed by POCh (0.037 mL, 0.400 mmol) in dropwise. The reaction mixture was stirred for 1 hour at ambient temperature, then concentrated in vacuo, and the e was ed by column chromatography DCM=0:10-1:10) to give the title comound (120 mg, 0.084 mmol, 42.2 % yield) as a semi-solid. LCMS (Method m, Table 7) R1 2.065 min, m/z 1422 (M+H-100).
Step 3: Synthesis of l-amino-N-(4-(4-((2S,6aS,6bR,7S,8aS,8bS,llaR,12aS,12bS)-2,6b­ Difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, 12a, 12bdodecahydro-1H-naphtho [2', l ':4,5]indeno[l,2-d][l,3]dioxol-l0-yl)phenoxy)phenyl)- 3,6,9,12, 15, 18,21,24,27,30,33,36-dodecaoxanonatriacontanamide To a stirred solution of tert-butyl (39-(( 4-( 4-( (2S,6aS,6bR,7S,8aS,8bS, 1OR l laR,12aS, l2bS)-8b-(2-((tert-butyldimethylsilyl)oxy)acetyl)-2,6b-difluorohydroxy -6a,8a-dimethyloxo- 2, 4, 6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5] indeno[l,2-d][l,3]dioxol-l 0- yl)phenoxy)phenyl)amino)oxo-3,6,9,12,15,18,21,24, 33,36-dodecaoxanonatriacontyl)carbamate (190 mg, 0.134 mmol) in methylene de (0.5 mL) was added TFA (0.1 mL, 1.298 mmol), and the solution was stirred for 2 hours at ambient temperature, then concentrated in vacuo to give the title compound (100 mg, 0.083 mmol, 62.0 % yield). LCMS (Method m, Table 7) R1 1.521 min, m/z 1208 (M+H).
Step 4: Synthesis of N-(4-( 4-((2S,6aS,6bR7S,8aS,8bS,1 OR l laR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro- hth o[2', l':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)phenoxy)phenyl)(3-(2,5-dioxo-2,5-dihydro-lH­ pyrrol- l-yl)propanamido)-3,6,9,12,15,18,21,24,27,30,33,36-dodecaoxanonatriacontanamide DIPEA (6.99 µL, 0.040 mmol) was added to a solution of l-amino-N-(4-(4- aS,6bR,7S,8aS,8bS, l laR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacet yl)-6a,8adimethyloxo-2 ,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho [2', l ':4,5]indeno[l,2- d][l,3]dioxol- l0-yl)phenoxy)phenyl)-3,6,9,12,15,18,21,24,27,30,33, 36-dodecaoxanonatriacontan amide (0.024 g, 0.02 mmol) and 2,5-dioxopyrrolidin-l-yl 3-(2,5-dioxo-2,5-dihydro-lH-pyrrol-lyl )propanoate (7.99 mg, 0.030 mmol) in N,N-dimethylformamide (1 mL), and the mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with EtOAc (10 mL) and washed with water (2xl0 mL), brine(lxl0 mL), dried over Na2SO4, filtered and concentratedin vacuo. The residue was purified by column chromatography (MeOH/DCM=0:100- 10:100) to give the title comound (0.011 g, 8.20 µmol, 41 % yield). LCMS (Method m, Table 7) Rt= 1.679 min, m/z 1359 (M+H). e 14: Synthesis of2,5-Dioxopyrrolidin- l-yl 6-(((S)- l-(((S)((3-(4- ((2S,6aS,6bR7S,8aS,8bS,1OR,llaR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyl -2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d] [l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)amino)oxopropanyl)amino) oxohexanoate (Cpd. No. 78) HO .. U� F+-{ 0 F 0 07 �HN r-f°o o )- l( N' �o,N)Z_ .,,0 NH ,, o L .• 0 0 µ cf' 1 0 0 ' 0 0 l 0 H O r-7) 0••' qo�f 'r � OH "J ( � A solution of (S)amino-N-((S)- l-((3-(4-((2S,6aS,6bR7S,8aS,8bS,l0R, l laR,12aS,12bS)- 2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dime thyloxo-2,4,6a,6b,7,8,8a,8b,l la,12,12a,12bdodecahydro-1H-naphtho [2', l ':4,5]indeno[l,2-d][l,3]dioxol- benzyl)phenyl)amino)oxopropan yl)propanamide (0.060 g, 0.080 mmol) and N,N-diisopropylethylamine (0.14 mL, 0.802 mmol) in DMSO (1 mL) was added ise to a room temperature solution of bis(2,5-dioxopyrrolidin- l-yl) adipate (0.273 g, 0.802 mmol) in DMSO (3.5 mL). After 60 min the reaction was quenched by addition of a 7 wt% solution of TFA in water to bring the reaction mixture to a pH of 4-5. The crude reaction mixture was ed by reverse phase HPLC on a Phenomenex Cl8(2) 5 micron column (250 x 21 mm column).
A gradient of MeCN (A) and 0.1% formic acid in water (B) was used, at a flow rate of30 mL/min (0-1.0 min 15% A, 1.0-11 min linear gradient 15-80% A, hold 1 min). Combined fractions were concentrated under reduced pressure to remove volatile solvents, and the resulting solution was frozen and lyophilized to give the title compound as a white solid (21.2 mg, 0.022 mmol, 27% yield). LCMS (Method r, Table 7) R1 = 0.80 min, m/z = 1005.1 [M+MeOH+H+]. 1HNMR (DMSO) 8 0.84 (s, 3H), 1.17 (d, J= 7. lHz, 3H), 1.25 (d, J= 7. lHz, 3H), 1.48 (s, 4H), 1.57 (q, J= 6.2Hz, 4H), 1.68 (dq, J= 13.7, 6.3Hz, 3H), 1.99-2.06 (m, lH), 2.09-2.18 (m, 2H), 2.18-2.36 (m, 2H), 2.55-2.72 (m, 3H), 2.78 (s, 4H), 3.87 (s, 2H), 4.14-4.22 (m, 2H), 4.26 (p, J= 7. lHz, lH), 4.33 (p, J= 7. lHz, lH), 4.49 (d, J= 19.4Hz, lH), 4.93 (d, J= 5.lHz, lH), 5.43 (s, lH), 5.49 (d, J= 5.4Hz, lH), 5.54-5.75 (m, lH), 6.11 (s, lH), 6.28 (dd, J= 10.2, 2.0Hz, lH), 6.89 (d, J= 7.6Hz, lH), 7.17 (t, J= 7.9Hz, lH), 7.23 (t, J= 9.7Hz, 3H), 7.34 (d, J= 7.8Hz, 2H), 7.39 (s, lH), 7.44 (d, J= 8.lHz, lH), 7.99 (d, J= 7.2Hz, lH), 8.02 (d, J= 7.3Hz, lH), 9.77 (s, lH); MS (ESI-) m/z = 971.
Example 15: sis of tert-butyl ((S)- l-(((S)((3-(4- ((2S,6aS,6bR7S,8aS,8bS,1 OR, l laR, 12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyloxo-2 ,4,6a,6b,7,8,8a,8b, l la, 12, 12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d] [l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)amino)methyl- l-oxobutan yl)carbamate BoeValAlaOH HATU, 2,6lut, THF o••' 0 Bo,HXr -d OH �0�,Qj) 6] HATU (106 mg, 0.280 mmol) and 2,6-lutidine (0.1 mL, 0.859 mmol) were added to a room temperature suspens10n of (2S,6aS,6bR, 7S,8aS,8bS, 1 OR, l laR, 12aS, 12bS)-l 0-( 4-(3- aminobenzyl)phenyl)-2,6b-difluorohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- 6a,6b,7,8,8a,8b, l la,12,12a,12b-decahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one (113 mg, 0.187 mmol) and (tert-butoxycarbonyl)-L-valyl-L-alanine (53.8 mg, 0.187 mmol) in THF (1.25 mL).
After 8h the reaction was diluted with EtOAc (16 mL), then washed sequentially with a IM aqueous on ofHCl (4 mL x 3), a saturated aqueous solution ofNaHCO3 (4 mL), and then a saturated s solution of brine (4 mL). t was removed under reduced pressure and the product was purified by chromatography (12 g silica), eluting with a gradient of 0-10% MeOH/DC M to give the title compound (148.6 mg, 0.170 mmol, 91% yield). LCMS (Method r, Table 7) R1 = 0.94 min, m/z = 875.9 [M+H+]. 1H NMR (DMSO-d6) 8 9.85 (s, lH), 7.99 (d, J = 7.1 Hz, lH), 7.43 (dd, J = 8.0, 1.7 Hz, lH), 7.36 - 7.31 (m, 3H), 7.27 - 7.15 (m, 5H), 6.89 (d, J = 7.5 Hz, lH), 6.67 (d, J = 8.8 Hz, lH), 6.27 (dd, J = 10.2, 1.9 Hz, lH), 6.11 (s, lH), 5.73 - 5.52 (m, lH), 5.50 (dd, J = 4.5, 1.7 Hz, lH), 5.43 (s, lH), 5.07 (t, J = 5.9 Hz, lH), 4.93 (d, J = 4.8 Hz, lH), 4.49 (dd, J = 19.5, 6.4 Hz, lH), 4.37 (t, J = 7.0 Hz, lH), 4.25 -4.12 (m, 2H), 3.87 (s, 2H), 3.80 (t, J = 7.7 Hz, lH), 2.73 - 2.53 (m, lH), 2.23 (ddd, J = 18.7, 11.9, 6.0 Hz, 2H), 2.08 - 1.99 (m, lH), 1.93 (q, J = 7.0 Hz, lH), 1.77 - 1.59 (m, 3H), 1.48 (s, 3H), 1.35 (s, 9H), 1.25 (d, J = 7.0 Hz, 3H), 0.89 -0.74 (m, 9H). e 16: Synthesis of S,6bR,7S,8aS,8bS, l0R, l laR,12aS,12bS)(4-((4- Aminophenyl)thio)phenyl)-2,6b-difluorohydroxy -8b-(2-hydroxyacetyl)-6a,8a,1 0-trimethyl- 6a,6b,7,8,8a,8b, l la,12,12a,12b-decahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one and (2S,6aS,6bR,7S,8aS,8bS, lOS, l laR,12a5,12bS)-l 0-( 4-((4-Aminophenyl)thio )phenyl)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a, 1 0-trimethyl-6a,6b,7,8,8a,8b, l la, 12, 12a,12b-decahydro-1H­ naphtho[2', l ':4,5]indeno[1,2-d] [ l,3]dioxol-4(2H)-one Step 1: Synthesis of 1-(4-((4-bromophenyl)thio)phenyl)ethanone K2CO3 DMF, ..
S �Br 8] 1-(4-Fluorophenyl)ethanone (2.19 mL, 18.04 mmol) was added to a stirred solution of 4- bromobenzenethiol (3.1 g, 16.40 mmol) and K2CO3 (2.72 g, 19.67 mmol) in DMF (45 mL), whereupon the reaction was heated to 100 °C for 20 min. The reaction was cooled to ambient temperature, diluted with water (50 mL) and extracted with EtOAc (3 X 50 mL). The combined organics were dried (MgSO4) and ts were removed under reduced pressure. cation by chromatography (silica, 120 g) eluting with a gradient of 0-60% EtOAc/heptanes gave the title compound (3.24 g, 10.55 mmol, 64% yield) as a yellow solid. LCMS (Method r, Table 7) Rt 0.95 min; m/z 307.0 [M+H+]. 1H NMR (400 MHz, DMSO-d6) 8 7.87 (d, J 8.7 Hz, 2H), 7.62 (d, J 8.6 Hz, 2H), 7.38 (d, J 8.6 Hz, 2H), 7.28 (d, J 8.7 Hz, 2H), 2.50 (s, 3H).
Step 2: Synthesis of tert-butyl ( 4-((4-acetylphenyl)thio)phenyl)carbamate I-BuOCONH2 XPhos, Pd2dba3 Cs2CO3 0PS�� NHBoc Nitrogen was sparged through a mixture of 1-(4-((4-bromophenyl)thio)phenyl)ethanone (3.24 g, 10.55 mmol), tert-butyl ate (1.483 g, 12.66 mmol), Cs2CO3 (5.15 g, 15.82 mmol), and dicyclohexyl(2',4',6'-triisopropyl-[l,l'-biphenyl]yl)phosphine (0.503 g, 1.055 mmol) in oxane for min. The flask was evacuated and back filled with N2 (3X). Pd2dba3 (0.290 g, 0.316 mmol) was added and the reaction was evacuated and back filled with N2 (3X). The reaction mixture was heated to 100 °C for 18h. The reaction was cooled to ambient temperature, treated with water (75 mL), then extracted with EtOAc (3 X 50 mL), dried (MgSO4), and solvents were removed under reduced pressure. Purification by chromatography a, 120 g) eluting with a nt of 0-60% EtOAc/heptanes gave the title compound (2.0 g, 5.82 mmol, 55% yield) as a yellow solid. LCMS (Method r, Table 7) Rt 0.96 min; m/z 344.0 [M+H+]. 1H NMR (501 MHz, DMSO-d6) 8 9.62 (s, lH), 7.82 (d, J 8.7 Hz, 2H), 7.58 (d, J 8.7 Hz, 2H), 7.43 (d, J 6.7 Hz, 2H), 7.11 (d, J 8.7 Hz, 2H), 2.49 (s, 3H), 1.47 (s, 9H).
Step 3: Synthesis of (2S,6aS,6bR,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((4- Aminophenyl)thio)phenyl)-2,6b-difluorohydroxy -8b-(2-hydroxyacetyl)-6a,8a,10-trimethyl- 6a,6b,7,8,8a,8b,I la, 12, 12a,12b-decahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one and S,6bR,7S,8aS,8bS,IOS, l a5,12bS)-l 0-(4-((4-Aminophenyl)thio)phenyl)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a,I0-trimethyl-6a,6b,7,8,8a,8b,I la, 12, 12a,12b-decahydro-1H­ naphtho[2', I': 4,5]indeno[ 1,2-d][ l,3]dioxol-4(2H)-one OH (\(S--A O 0� lANH2 ... o F F Triflic acid (0.431 mL, 4.85 mmol) was added drop-wise to a 0 °C slurry of ,9R,10S,I IS,13S,14S,16R,17S)-6,9-difluoro- l l,16,17-trihydroxy(2-hydroxyacetyl)-10,13- dimethyl-6,7,8,9,10,11,12,13,14,15,16, l 7-dodecahydro-3H-cyclopenta[a]phenanthrenone (0.400 g, 0.970 mmol), tert-butyl (4-((4-acetylphenyl)thio)phenyl)carbamate (0.366 g, 1.067 mmol), and MgSO4 (0.350 g, 2.91 mmol) in MeCN (4.0 mL). After 30 min the reaction was diluted with EtOAc (25 mL), washed sequentially with a saturated aqueous solution of NaHCO 3 (20 mL), with a saturated aqueous solution ofbrine (25 mL), dried (MgSO4), and then solvent was removed under reduced pressure to give a yellow foam. Purification by chromatography (silica, 40 g) eluting with a gradient of 0-10% MeOH/DCM gave the product as a mixture of the ketal isomers (460 mg, 0.721 mmol, 74% yield). A n of this material was purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 X 50 mm). A gradient of MeCN (A) and 0.1 % TFA in water (B) was used, at a flow rate of 90 mL/min (0-5.0 min 15% A, 5.0-18 min linear gradient 15-75% A, then hold 5 min). Combined fractions were concentrated to remove volatile solvents under reduced re, and the resulting solutions were frozen and lyophilized to give the ketal isomers as yellow . Minor ketal isomer: (2S,6a5,6bR,7S,8aS,8bS,IOR,11aR, l 2a5, l 2bS)-l 0-(4-((4-aminophenyl)thio)phenyl)-2,6b-difluoro- 7- hydroxy-8b-(2-hydroxyacetyl)-6a,8a,I0-trimethyl-6a,6b,7,8,8a,8b,I la, 12, 12a,12b-decahydro-1H­ naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one. Yellow powder (10.0 mg). LCMS (Method r, Table 7) Rt 0.80 min; m/z 638.2 [M+H+]. 1H NMR (500 MHz, DMSO-d 8.5 Hz, 2H), 6) 8 7.33 (d, J 7.23 (dd, J= IO.I, 1.5 Hz, IH), 7.19 -7.12 (m, 2H), 6.96 (d, J= 8.5 Hz, 2H), 6.76 - 6.63 (m, 2H), 6.29 (dd, J I0.2, 1.9 Hz, IH), 6.10 (s, IH), 5.66 -5.45 (m, 2H), 5.14 (d, J 5.8 Hz, IH), 4.65 (d, J 19.3 Hz, IH), 4.22 - 4.07 (m, 2H), 2.48 -2.35 (m, IH), 2.14 - 2.04 (m, IH), 2.02 -1.91 (m, IH), 1.77 -1.64 (m, 2H), 1.63 -1.56 (m, IH), 1.50 (dd, J 13.2, 6.3 Hz, IH), 1.44 (s, 3H), 1.36 (s, 3H), 1.14 -0.98 (m, IH), 0.80 (s, 3H). Major ketal isomer: (2S,6aS,6bR,7S,8aS,8bS,l0S,llaR,12aS,12bS)(4-((4- aminophenyl)thio)phenyl)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a,I 0-trimethyl- 6a,6b,7,8,8a,8b,I 12a,12b-decahydro-1H-naphtho[2', ]indeno[l,2-d][l,3]dioxol-4(2H)-one.
Yellow powder (18.1 mg). LCMS (Method r, Table 7) Rt 0.85 min; m/z 638.2 [M+H+]. 1H NMR (500 MHz, DMSO-d6) 8 7.28 (d, J I0.2 Hz, IH), 7.20 (dd, J 8.4, 7.2 Hz, 4H), 6.95 (d, J 8.4 Hz, 2H), 6.73 (d, J= 8.5 Hz, 2H), 6.31 (d, J= 12.0 Hz, lH), 6.13 (s, lH), 5.75 5.57 (m, lH), 5.53 (s, lH), 5.00 (d, J 5.1 Hz, lH), 4.22 (d, J 7.2 Hz, lH), 4.06 3.80 (m, 4H), 2.72 2.55 (m, lH), 2.39 2.27 (m, lH), 2.17 2.02 (m, 2H), 1.79 1.56 (m, 3H), 1.50 (d, J 12.4 Hz, 6H), 0.73 (s, 3H).
Example 17: Synthesis of(6aR,6bS,7S,8aS,8bS, l0R, l laR,12aS,12bS)(4-((4- henyl)thio)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a,10-trimethyl- 6a,6b,7,8,8a,8b, l la,12, 12a,12b-decahydro-1H-naphtho[2', l':4,5]indeno[ l,2-d][l,3]dioxol-4(2H)-one and ( 6aR6bS,7S,8aS,8bS,1 OS, l laR,12aS,12bS)- l0-( 4-(( ophenyl)thio)phenyl)hydroxy-8b-(2- hydroxyacetyl)-6a,8a,10-trimethyl -6a,6b,7,8,8a,8b, l la,12, 12a,12b-decahydro-1H- naphtho[2', l':4,5]indeno[1,2-d] [ l,3]dioxol-4(2H)-one 3] Triflic acid (0.24 mL, 2.66 mmol) was added drop-wise to a 0 °C slurry of (8S,9S,10R,11S,13S,14S,16R, l7S)- l l,16,17-trihydroxy(2-hydroxyacetyl)-10,13-dimethyl- 6,7,8,9,10, l 3,14,15,16,l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (0.200 g, 0.531 mmol), tert-butyl (4-((4-acetylphenyl)thio)phenyl)carbamate (0.201 g, 0.584 mmol), and MgSO4 (0.192 g, 1.59 mmol) in MeCN (2.0 mL). After 30 min the reaction was d with EtOAc (15 mL), washed sequentially with a saturated aqueous solution of NaHCO3 (10 mL), and then with a saturated aqueous solution of brine (10 mL), dried (MgSO4), and solvent was removed under reduced pressure to give a yellow foam. Purification by chromatography (silica, 24 g) eluting with a gradient of 0-10% MeOH/DCM gave the product as a e of the ketal isomers (198 mg, 0.329 mmol, 62% yield). A portion of this al was purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 X 50 mm). A gradient of MeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of 90 mL/min (0-5.0 min 15% A, 5.0-18 min linear gradient 15-75% A, then hold 5 min). Combined ons were concentrated to remove volatile solvents under reduced pressure, and the resulting solution was frozen and lyophilized to give both ketal isomers as white solids. Major ketal isomer: (6aR,6bS,7S,8aS,8bS, lOS, l laR,12aS,12bS)- l0-(4-((4-aminophenyl)thio)phenyl)hydroxy-8b-(2- hydroxyacetyl)-6a,8a,10-trimethyl-6a,6b,7,8,8a,8b, l la,12, 12a,12b-decahydro-1H- naphtho[2', l':4,5]indeno[ l,2-d][l,3]dioxol-4(2H)-one. White powder (14.6 mg). LCMS (Method r; Table 7) R1 0.83 min; m/z 602.1 [M+H+]. 1H NMR (501 MHz, DMSO-d6) 8 7.30 (d, J= 10.1 Hz, lH), 7.22 - 7.12 (m, 4H), 6.91 (d, J= 8.5 Hz, 2H), 6.68 (d, J= 8.5 Hz, 2H), 6.16 (dd, J= 10.1, 1.9 Hz, lH), 5.91 (s, lH), 4.93 (d, J 4.6 Hz, lH), 4.74 (hrs, 2H), 4.30 (d, J 2.9 Hz, lH), 4.02 - 3.79 (m, 4H), 2.53 (dt, J= 14.7, 7.6 Hz, lH), 2.30 (d, J= 14.8 Hz, lH), 2.16 -1.95 (m, 2H), 1.85 (d, J= 3.6 Hz, lH), 1.78 - 1.67 (m, 2H), 1.55 (td, J 15.2, 13.3, 7.7 Hz, 2H), 1.50 (s, 3H), 1.37 (s, 3H), 1.13 -0.97 (m, 2H), 0.70 (s, 3H). Minor ketal isomer: (6aR,6bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((4- aminophenyl)thio)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a,1 0-trimethyl- 6a,6b,7,8,8a,8b, l la,12,12a,12b-decahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one.
White powder (12.0 mg). LCMS (Method r, Table 7) Rt 0.80 min; m/z 602.l[M+H+]. 1HNMR (501 MHz, DMSO-d6) 8 7.32 (d,J 6.8 Hz, 2H), 7.25 (d,J 10.1 Hz, lH), 7.13 (d,J 8.5 Hz, 2H), 6.95 (d,J 8.5 Hz, 2H), 6.66 (d, J= 8.5 Hz, 2H), 6.13 (dd, J= 10.1, 1.9 Hz, lH), 5.87 (s, lH), 5.09 (d, J= 6.1 Hz, lH), 4.71 (hrs, lH), 4.62 (d, J 19.3 Hz, lH), 4.22 (d, J 2.9 Hz, lH), 4.11 (d, J 19.2 Hz, 2H), 2.47 - 2.37 (m, lH), 2.25 - 2.07 (m, lH), 1.94 (qd, J 11.3, 3.8 Hz, lH), 1.87 -1.75 (m, lH), 1.70 (s, 2H), 1.59 - 1.44 (m, 2H), 1.32 (d, J 5.1 Hz, 6H), 1.18 -1.03 (m, lH), 0.78 (s, 3H), 0.61 (dd, J 11.2, 3.5 Hz, lH), 0.50 (qd, J 12.9, 4.8 Hz, lH).
Example 18: Synthesis of2S,6aS,6bR,7S,8aS,8bS,l0R,l laR, 12aS,12bS)(4-(4-Aminophenoxy) hydroxyphenyl)-2,6b-difluorohydroxy-8b-(2-hydroxyac 6a,8a-dimethyl - 6a, 6b,7,8,8a,8b, l la, 12,12a,12b-decahydro-1H-naphth o[2', l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one Step 1: Synthesis of 3-Methoxy(4-nitrophenoxy)benzaldehyde ] Vanillin (2.5 g, 16.43 mmol), 4-Fluoronitrobenzene (2.61 mL, 24.65 mmol), and potassium carbonate (4.54 g, 32.9 mmol) were dissolved in DMF (15 mL) and d at 80 °c overnight. After cooling, the mixture was treated with water, and extracted with EtOAc (x2). The combined organic layers were washed with water and brine, dried (Na 2SO4), and concentrated. Purification by tography (silica, 120 g) eluting with a gradient of 0-40% EtOAc in heptanes afforded the title compound as a slightly yellow solid (3.37 g, 75%). LCMS d r, Table 7) Rt 0.88 min; m/z not observed. 1H NMR(400 MHz, DMSO-d6) 810.00 (s, lH), 8.26 -8.17 (m, 2H), 7.72 - 7.60 (m, 2H), 7.42 (d,J 8.1 Hz, lH), 7.12 -7 .03 (m, 2H), 3.82 (s, 3H).
Step 2: Synthesis of 3-hydroxy(4-nitrophenoxy)benzaldehyde Tribromoborane (110 mL, 110 mmol) was added to a -78 °C solution of 3-methoxy(4- nitrophenoxy)benzaldehyde (6.02 g, 22.03 mmol) in DCM (100 mL). The reaction was stirred at -78 °C for 1 h, then stirred at 0 °C for 5 hours. The mixture was mixed with ice, and ted with DCM. The combined organic layers were washed with water and brine, dried (Na2SO4), and concentrated.
Purification by chromatography (silica, 120 g) eluting with a gradient of 0-30% EtOAc in heptanes afforded the title nd as a purplish oil (5.55 g, 97% yield). LCMS (Method r, Table 7) Rt 0.80 min; m/z not observed. 1H NMR (400 MHz, 6) 8 10.24 (s, lH), 8.22 -8.14 (m, 2H), 7.36 (s, lH), 7.30 (d,J 2.1Hz, lH), 7.15-7.07 (m, 2H), 7.02-6.96 (m, 2H).
Step 3: Synthesis of4-(4-aminophenoxy)hydroxybenzaldehyde Stannous chloride (18.29 g, 96 mmol) was added to a solution of added oxy(4- nitrophenoxy)benzaldehyde (5.g, 19.29 mmol), stannous chloride (18.29 g, 96 mmol) in ethanol (60 mL), which was heated to 80 °C for 2h. The mixture was cooled and mixed carefully with ice, and saturated sodium bicarbonate aqueous solution, then extracted with EtOAc multiple times. The combined organic layers were washed with brine, dried (Na ®, and the filtrate was concentrated 2SO4), filtered through Celite to afford the title compound as a yellow solid (1.18 g, 27% yield). LCMS (Method r, Table 7) Rt 0.48 min; m/z not observed. 1H NMR (400 MHz, DMSO-d6) 8 9.90-9.87 (m, lH), 10.90-9.26 (m, 2H), 8.66-8.56 (m, lH), 7.66-7.61 (m, lH), .46 (m, lH), 7.46 -7.38 (m, 2H), 7.38 -7.29 (m, 4H), 7.16 -6.99 (m, 6H); MS(ESI-) m/z 227.9 (M-H).
Step 4: sis of (2S,6aS,6bR,7S,8aS,8bS,10R, l laR,12aS,12bS)( 4-( 4- Aminophenoxy)hydroxyphenyl)-2, 6b-difluorohydroxy -hydroxyacetyl)-6a, ethyl - 6a,6b,7,8,8a,8b, l la, 12,12a,12b-decahydro-1H-naphtho[2', l ':4,5]indeno[ l,2-d][l,3]dioxol-4(2H)-one Step4 0 Perchloric acid (2.64 mL, 24.25 mmol) was added to a room temperature solution of 4-(4- aminophenoxy)hydroxybenzaldehyde (0.611 g, 2.67 mmol) and ,9R l0S, l1S,13S,14S,16R,l7S)-6,9-difluoro- l l,16, l 7-trihydroxy-l7-(2-hydroxyacetyl)-10,13- dimethyl-6,7,8,9,10,11,12,13,14,15,16,l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (1 g, 2.425 mmol) in THF (70 mL). After 16 hours the reaction was treated with water and extracted twice with EtOAc. The combined organic layers were washed with a saturated s solution of sodium bicarbonate, a saturated aqueous solution of sodium thiosulfate solution, then a saturated aqueous solution of brine, dried (Na 2SO4), and solvent was removed under reduced pressure. The al was purified by reverse phase HPLC on a Phenomenex Cl8(2) 5 micron column (250 X 21 mm column). A gradient of MeCN (A) and 0.1% TFA in water (B) was used at a flow rated of30 mL/min (0.0-1.0 min 15% A, 1.0-10 min linear gradient to 65% A, hold 1 min). Combined fractions were concentrated under reduced pressure to remove volatile solvents, and the resulting solution frozen and lyophilized to give the title t as a yellow solid (338.9 mg, 23% . LCMS (Method r, Table 7) Rt 0.72 min; MS (ESI+) 624.2 (M+H); 1H NMR (400 MHz, DMSO-d6) 8 9.88 (s, 2H), 7.28 (dd, lH), 7.27 7.22 (m, 2H), 7.07 (d, lH), 7.00 (d, lH), 6.96 6.88 (m, 3H), 6.30 (dd, lH), 6.18 6.08 (m, lH), 5.78 5.67 (m, lH), 5.65 5.52 (m, lH), .42 (s, lH), 5.00 4.95 (m, lH), 4.53 (d, lH), 4.27 4.18 (m, 2H), 2.79 2.57 (m, lH), 2.36 2.28 (m, lH), 2.24 (td, lH), 2.13 2.01 (m, lH), 1.80 1.66 (m, 3H), 1.65 1.52 (m, lH), 1.51 (s, 3H), 0.88 (s, Example 19: Synthesis of (6aR,6bS,7S,8aS,8bS,l0R,l l aR,12aS,12bS)(4-((4- Aminophenyl)sulfonyl)pheny1)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- 6a, 6b, 7,8,8a,8b, l 12a,12b-decahydro-1H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one Step 1: Synthesis of 4-((4-Bromophenyl)thio)benzonitrile SH F K2C03 r(y + � DMF, 120 °C Br� �CN 3] Potassium carbonate (4.39 g, 31.7 mmol) was added to a solution of 4-bromobenzenethiol (5.0 g, 26.4 mmol) and 4-fluorobenzonitrile (3.20 g, 26.4 mmol) in DMF (50 mL), which was heated to 120 °C for 3h. The reaction was cooled to 0 °C water was added (100 mL) and the mixture was extracted with EtOAc (3 X 50 mL). The ed organics were dried (MgSO4) and solvent was removed under reduced pressure. Purification by chromatography (80 g silica) eluting with a gradient of 0-60% heptanes gave the title compound (6.82 g, 23.5 mmol, 89% yield) as a yellow solid. LCMS (Method r, Table 7) Rt 0.95 min; m/z 291.2 . 1H NMR (400 MHz, DMSO-d6) 8 7.72 (d, J 8.7 Hz, 2H), 7.65 (d, J 8.5 Hz, 2H), 7.43 (d, J 8.5 Hz, 2H), 7.26 (d, J 8.7 Hz, 2H).
Step 2: Synthesis of tert-butyl (4-((4-cyanophenyl)thio)phenyl)carbamate tBuOCONH2 XPhos, Pd2dba3 Cs2C03 Nitrogen was sparged through a mixture of 4-((4-bromophenyl)thio)benzonitrile (6.0 g, 20.68 mmol), tert-butyl carbamate (2.91 g, 24.81 mmol), diisopropyl(2',4',5'-triisopropyl-[l,1'-biphenyl] yl)phosphine (0.820 g, 2.068 mmol), and Cs2CO3 (10.11 g, 31.0 mmol) in 1,4-dioxane (207 mL) for 30 min. The flask was evacuated and back filled with N2 (3X). Pd2dba3 (0.568 g, 0.620 mmol) was added and the reaction was evacuated and back filled with N2 (3X) times. The reaction e was heated to 100 °C for 28h. The reaction was cooled to room temperature, pon it was treated with water (200 mL), extracted with EtOAc (3 X 75 mL), dried (MgSO4) and solvents were removed under reduced pressure. Purification by chromatography (silica, 120 g) eluting with a gradient of 0-30% EtOAc/heptanes gave the title nd (3.20 g, 9.80 mmol, 47% yield) as a yellow solid. LCMS (Method r, Table 7) Rt 1.0 min; m/z 344.1 [M+NH/]. 1H NMR (400 MHz, DMSO-d6) 8 9.67 (s, lH), 7.69 (d, J 8.7 Hz, 2H), 7.61 (d, J 8.7 Hz, 2H), 7.47 (d, J 8.7 Hz, 2H), 7.14 (d, J 8.6 Hz, 2H), 1.49 (s, 9H).
Step 3: Synthesis oftert-butyl (4-((4-cyanophenyl)sulfonyl)phenyl)carbamate 0 0,, ,, m CPBA 8 CH2Cl2 NC0 0NHBoc 3-Chloroperoxybenzoic acid (639 mg, 3.71 mmol) was added n-wise to a room temperature solution of tert-butyl (4-((4-cyanophenyl)thio)phenyl)carbamate (480 mg, 1.471 mmol) in CH2Ch (15 mL). After 30 min, the on was portioned between water (20 mL) and EtOAc (10 mL).
The layers were separated and the aqueous phase was extracted with EtOAc (2 X 25 mL). The ed organics were washed with a saturated aqueous solution of brine (50 mL), dried over MgSO4, and solvents were removed under reduced re. Purification by chromatography (silica, 40 g) eluting with a gradient of 0-60% EtOAc/heptanes gave the title compound (372 mg, 1.04 mmol, 71 % yield) as a yellow solid. LCMS (Method r, Table 7) Rt 0.86 min; m/z 376.0 [M+NH/]. 1H NMR (400 MHz, DMSO-d6) 8 9.92 (s, lH), 8.06 (s, 4H), 7.87 (d, J= 8.9 Hz, 2H), 7.66 (d, J= 9.0 Hz, 2H), 1.45 (s, 9H).
Step 4: Synthesis oftert-butyl (4-((4-formylphenyl)sulfonyl)phenyl)carbamate 0 0,, ,, DiBAIH; 8 1N aq. HCI OHC0 0NHBoc Diisobutylaluminum hydride (6.53 mL, 1.0 M in toluene, 6.53 mmol) was added drop-wise over 5 minutes to a 0 °C on of utyl (4-((4-cyanophenyl)sulfonyl)phenyl)carbamate (0.780 g, 2.176 mmol) in THF (20 mL). After 30 min diisobutylaluminum hydride (1.0 Min toluene) (2.176 mL, 2.176 mmol) was added and the reaction was stirred at 0 °C for an additional lh. The reaction was quenched at 0 °C by slow addition ofal N aqueous solution ofHCl (120 mL) and the aqueous phase was ted with EtOAc (2 X 75 mL). The combined organics were washed with a saturated aqueous solution of brine (50 mL), dried over MgSO4 and solvents were removed under reduced pressure.
Purification by tography (silica, 80 g) eluting with a gradient of0-10% CH2Ch/MeOH gave the title compound (0.275 g, 0.761 mmol, 35% yield) as a yellow oil. LCMS (Method r, Table 7) R1 = 0.83 min; m/z = 359.9 [M-f]. 1H NMR (400 MHz, DMSO-d6) 8 10.04 (s, lH), 9.89 (s, lH), 8.18 - 7.97 (m, 4H), 7.85 (d, J= 8.9 Hz, 2H), 7.64 (d, J= 8.9 Hz, 2H), 1.43 (s, 9H).
Step 5: Synthesis of (6aR,6bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((4- Aminophenyl)sulfonyl)pheny1)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- 6a,6b,7,8,8a,8b,l la, 12,12a,12b-decahydro-1H-naphtho[2',l ':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one TfOH, MgSO4 ACN, 0-5 °C Triflic acid (0.12 mL, 1,328 mmol) was added drop-wise to a 0 °C slurry of (8S,9S,10R, l lS,13S,14S,16R,17S)-l l,16,17-trihydroxy(2-hydroxyacetyl)-10,13-dimethyl- 6,7,8,9,10, l l,12,13,14,15,16, l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (0.100 g, 0.266 mmol), utyl (4-((4-formylphenyl)sulfonyl)phenyl)carbamate (0.106 g, 0.292 mmol), and MgSO4 (0.096 g, 0.797 mmol) in MeCN (1.0 mL). After 30 minutes the reaction was diluted with EtOAc (15 mL), and then washed with a saturated aqueous solution of NaHCO 3 (10 mL) followed by a saturated aqueous solution of brine (10 mL), and dried ). Removal of solvent under reduced re gave a light yellow foam, which was ed by chromatography (silica, 24 g) eluting with a gradient of 0-10% CH2Ch/MeOH to give a colorless glass. The acetal isomers were separated by preparative reverse phase HPLC on a enex Cl8 (2) 10 micron column, (250 X 30 mm). A gradient of MeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of 60 mL/min (0-3.0 min 15% A, 3.0-18 min linear gradient 15- 80% A, then hold 5 min). ed fractions were concentrated to remove volatile solvents under reduced pressure, and the resulting solution was frozen and lyophilized to give the title compound as a white solid (8.0 mg, 18% yield). LCMS (Method r, Table 7) R1 = 0.76 min; MS m/z = 620.0 [M+H+]. 1H NMR (400 MHz, DMSO-d6) 8 7.81 (d, J 8.4 Hz, 2H), 7.61 (d, J 8.4 Hz, 2H), 7.49 (d, J 8.8 Hz, 2H), 7.27 (d, J 10.0Hz, lH), 6.56 (d, J 8.8 Hz, 2H), 6.12 (dd, J 10.1, 1.9 Hz, lH), 5.89 (s, lH), 5.47 (s, lH), 4.91 (d, J 4.6 Hz, lH), 4.73 (s, lH), 4.48 (d, J 19.4Hz, lH), 4.24 (s, lH), 4.13 (d, J 19.5Hz, lH), 2.51 (s, 2H), 2.32 - 2.22 (m, lH), 2.13 -2.01 (m, lH), 2.02 -1.88 (m, lH), 1.78 - 1.56 (m, 5H), 1.35 (s, 3H), 1.11 - 0.96 (m, 2H), 0.82 (s, 3H).
Example 20: N-(3-(4-((2S,6aS,6bR,7S,8aS,8bS, 1 OR, l laR, 12aS,12bS)-2,6b-Difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, 12a,12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[1,2-d][ l,3]dioxol- l 0-yl)benzyl)phenyl)(2-(2-(3-(2,5-dioxo-2,5-dihydro- lH­ pyrrol- l-yl)propanamido)ethoxy)ethoxy)propanamide Step 1: Synthesis of tert-butyl (2-(2-(3-((3-(4-((2S,6aS,6bR,7S, 8aS,8bS,1 OR, l aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho[2',l ':4,5]indeno[ [l,3]dioxol-l 0- yl)benzyl)phenyl)amino)oxopropoxy)ethoxy)ethyl)carbamate HATU, 2,6-lul,, THF o ,,,- ,,,- ,,,lo BocHN_,,,..____,,_ ,,,,.._ o�NRO H,NRO � OH H OH HATU (0.125 g, 0.328 mmol) was added to a room temperature solution of 2,2-dimethyl oxo-3,8,11-trioxaazatetradecanoic acid (0.100 g, 0.361 mmol), S,6bR, ,8bS, 1OR, l laR,12aS,12bS)-l 0-( 4-(3-aminobenzyl)phenyl)-2,6b-difluorohydroxy- 8b-(2-hydroxyacetyl)-6a,8a-dimethyl-6a,6b,7,8,8a,8b, l la, 12,12a,12b-decahydro-1H- naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one (0.199 g, 0.328 mmol) and 2,6-dimethylpyridine (0.12 mL, 0.983 mmol) in THF (2.0 mL). After 24 hours solvents were removed under reduced pressure and the reaction mixture was purified by chromatography (silica, 24 g) eluting with a gradient of 0-10% MeOH/CH2Ch to give the title compound as a light yellow foam (226 mg, 0.261 mmol, 80% .
LC MS (Method r, Table 7) Rt= 0.91 min, m/z = 865.5 [M+H+].1H NMR (DMSO-d 6) 8 0.86 (s, 3H), 1.36 (s, 9H), 1.50 (s, 4H), 1.71 (ddt, J= 17.9, 13.3, 5.8 Hz, 3H), 1.94 -2.14 (m, 2H), 2.18- 2.39 (m, lH), 2.55 -2.74 (m, lH), 3.03 (q, J= 6.0 Hz, 2H), 3.48 (hept, J= 3.1, 2.7 Hz, 4H), 3.66 (t, J= 6.3 Hz, 2H), 3.88 (s, 2H), 4.13-4.26 (m, 2H), 4.51 (d, J= 19.4 Hz, lH), 4.94 (d, J= 5.1 Hz, lH), 5.45 (s, lH), 5.52 (dd, J= 4.3, 1.7 Hz, lH), 5.65 (dddd, J= 48.5, 11.4, 6.7, 2.0 Hz, lH), 6.13 (d, J= 2.1 Hz, lH), 6.73 (t, J= 5.8 Hz, lH), 6.80 -6.97 (m, lH), 7.18 (t, J= 7.8 Hz, lH), 7.25 (td, J= 9.1, 8.2, 1.6 Hz, 3H), 7.32 - 7.39 (m, 3H), 7.45 (dd,J= 8.4, 2.0 Hz, lH), 7.63 (d,J= 7.8 Hz, lH), 8.11- 8.85 (m, lH), 9.83 (s, lH).
Step 2: 3-(2-(2-Aminoethoxy)ethoxy)-N-(3-( 4-((2S,6aS,6bR, 7S,8aS,8bS, 1OR, l laR, l2aS, l2bS)-2,6b-difluorohydroxy- 8b-(2-hydroxyacetyl)-6a,8a-dimethy1oxo-2, 4,6a,6b, 7 ,8,8a, 8b, l la, 12, 12a, 12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol yl)benzyl)phenyl)propanamide � 0 TFA, THF o ,,,- ,,,- ...,lo BocHN_,,,..____,,_ o...._.,,,,.._O�N� H OH TFA (1.0 mL, 12.98 mmol) was added to a room temperature solution of tert-butyl (3- ((3-(4-((2S,6aS,6bR,7S,8aS,8bS,1OR, l laR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)- 6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d][l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropoxy)ethoxy)ethyl)carbamate (226 mg, 0.261 mmol) in CH2Ch (3.0 mL). After 45 min volatiles were d under vacuum and the crude product was carried on to the next step without further purification, assuming 100% yield. LCMS (Method r, Table 7) R1 = 0.80 min, m/z = 765.4 [M+H+].
Step 3: Synthesis of N-(3-(4-((2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR, 12aS,12bS)-2,6b-Difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, 12a,12b-dodecahydro- 1H-naphtho[ 2', l':4,5]indeno[1,2-d][ l,3]dioxol- l0-yl)benzyl)phenyl)(2-(2-(3-(2,5-dioxo-2,5-dihydrolH-pyrrol-l-yl )propanamido)ethoxy)ethoxy)propanamide N,N-Diisopropylethylamine (0.155 mL, 0.88 mmol) was added to a room temperature on of 3-(2-(2-aminoethoxy)ethox y)- N-(3-(4-((2S,6aS,6bR7S,8aS,8bS,l0R l laR12aS,12bS)-2,6bdifluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b, ,8b,l la, 12, 12a,12bdodecahydro-1H-naphtho [2',l':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)benzyl)phenyl)propanamide (0.226 g, 0.295 mmol) and 2,5-dioxopyrrolidin-l-yl 3-(2,5-dioxo-2,5-dihydro-lH-pyrrol- l-yl)propanoate (0.087 g, 0.325 mmol) in DMF (2.0 mL). After 45 min, the crude reaction mixture was purified by reverse phase HPLCon a Phenomenex Cl8(2) 10 micron column (250 x 50 mm column). A gradient of MeCN (A) and 0.1% formic acid in water (B) was used, at a flow rate of 80 mL/min (0-5.0 min 18% A, 5.0-25.0 min linear gradient 15-80% A, hold 5 min). Combined fractions were concentrated under d pressure to remove volatile solvents, and the ing solution was frozen and lyophilized to give the title compound as a white solid (48 mg, 0.052 mmol, 18% yield). LCMS (Method r, Table 7) R1 = 0.84 min, m/z = 916.4 [M+H+]. 1H NMR (DMSO-d6) 8 0.84 (s, 3H), 1.48 (s, 4H), 1.59 - 1.76 (m, 3H), 2.03 (d,J= 13.9 Hz, lH), 2.17 - 2.38 (m, 4H), 2.54 -2.72 (m, lH), 3.11 (q,J= 5.8 Hz, 2H), 3.31 - 3.35 (m, 4H), 3.42 - 3.51 (m, 4H), 3.57 (dd,J= 7.8, 6.8 Hz, 2H), 3.64 (t,J= 6.3 Hz, 2H), 3.86 (s, 2H), 4.10 - 4.25 (m, 2H), 4.49 (dd,J = 19.5, 6.0 Hz, lH), 4.93 (d,J= 5.1 Hz, lH), 5.07 (t,J= 5.9 Hz, lH), 5.43 (s, lH), 5.51 (s, lH), 5.53 - .74 (m, lH), 6.11 (s, lH), 6.28 (dd,J= 10.2, 1.9 Hz, lH), 6.88 (d,J= 7.5 Hz, lH), 6.97 (s, 2H), 7.16 (t,J = 7.8 Hz, lH), 7.20 - 7.28 (m, 3H), 7.30 - 7.39 (m, 3H), 7.38 - 7.48 (m, lH), 7.96 (t,J= 5.6 Hz, lH), 9.81 (s, lH).
Example 21: N-(3-(4-((2S,6aS,6bR,7S,8aS,8bS,1OR l laR,12aS,12bS)-2,6b-Difluorohydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la, 12, 12a,12b-dodecahydro-1H- o[2',l':4,5]indeno[l,2-d][l,3]dioxolyl)benzyl)phenyl)-l-(3-(2,5-dioxo-2,5-dihydro-1H-pyrroll-yl )propanamido)-3,6,9,12-tetraoxapentadecanamide Prepared by the same procedure as Example 20. White solid (17 mg, 0.017 mmol, 9% yield). LCMS d r, Table 7) Rt 0.82 min, m/z 1026 [M+Na+]. 1H NMR (DMSO-d 6) 8 0.85 (s, 3H), 1.22 (s, 8H), 1.49 (s, 3H), 1.61 - 1.77 (m, 2H), 2.03 (d,J 13.9 Hz, lH), 2.12 - 2.40 (m, 3H), 2.55 - 2.66 (m, lH), 3.12 (q,J 5.8 Hz, 2H), 3.33 (s, lH), 3.41 -3.51 (m, llH), 3.58 (t,J 7.3 Hz, 2H), 3.65 (t,J 6.3 Hz, 2H), 3.87 (s, 2H), 4.18 (d,J 14.1 Hz, 2H), 4.42 - 4.61 (m, lH), 4.93 (d,J 5.2 Hz, lH), 5.07 (s, lH), 5.44 (s, lH), 5.50 (s, lH), 5.6-5.7 (m, lH), 6.28 (dd,J 10.2, 1.9 Hz, lH), 6.88 (d,J 7.8 Hz, lH), 6.98 (s, 2H), 7.17 (t,J 7.9 Hz, lH), 7.24 (t,J 9.8 Hz, 3H), 7.32 - 7.38 (m, 3H), 7.43 (d,J 8.3 Hz, lH), 7.98 (s, lH), 9.81 (s, lH).
Example 22: N-(3-(4-((2S,6aS,6bR,7S,8aS,8bS,1 OR llaR12aS, 12bS)-2,6b-Difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4, 6a,6b,7,8,8a,8b,l la,12, 12a,12b-dodecahydro-1H- naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l 0-yl)benzyl)phenyl)(3-(2,5-dioxo-2,5-dihydro-lH-pyrroll-yl )propanamido)-3,6,9,12,15, l8-hexaoxahenicosanamide Prepared by the same procedure as Example 20. White solid (23.2 mg, 0.021 mmol, 22% yield). LCMS (Method r, Table 7) Rt 0.83 min, m/z 1092.3 . 1H NMR d 6) 8 0.84 (s, 3H), 1.48 (s, 4H), 1.58 - 1.76 (m, 3H), 2.02 (dt,J 14.0, 3.6 Hz, lH), 2.17 - 2.37 (m, 4H), 2.62 (<ltd,J 24.1, 11.9, 4.4 Hz, lH), 3.12 (q,J 5.8 Hz, 2H), 3.40 - 3.52 (m, 23H), 3.57 (t,J 7.3 Hz, 2H), 3.64 (t,J 6.3 Hz, 2H), 3.86 (s, 2H), 4.10 - 4.25 (m, 2H), 4.49 (d,J 19.4 Hz, lH), 4.92 (d,J 5.0 Hz, lH), 5.08 (s, lH), 5.43 (s, lH), 5.49 - 5.73 (m, 2H), 6.11 (s, lH), 6.27 (dd,J 10.1, 1.9 Hz, lH), 6.87 (d,J 7.6 Hz, lH), 6.97 (s, 2H), 7.16 (t,J 7.8 Hz, lH), 7.23 (dd,J 13.9, 9.0 Hz, 3H), 7.30 - 7.38 (m, 3H), 7.43 (d,J 8.1 Hz, lH), 7.98 (t,J 5.6Hz, lH), 9.81 (s, lH).
Example 23: N-(3-(4-((2S,6aS,6bR,7S,8aS,8bS,1 OR llaR12aS, 12bS)-2,6b-Difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4, 6a,6b,7,8,8a,8b,l la,12, 12a,12b-dodecahydro-1H- naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l enzyl)phenyl)(3-(2,5-dioxo-2,5-dihydro-lH-pyrroll-yl )propanamido)-3,6,9,12, 15, 18,21,24,27,30,33,36-dodecaoxanonatriacontanamide Prepared by the same procedure as Example 20. Isolated as a colorless glass (20 mg, 0.015 mmol, 18% yield). LCMS (Method r, Table 7) Rt= 0.85 min, m/z= 1356.4 [M+H+]. 1H NMR (DMSO-d 8 0.84 (s, 3H), 1.48 (s, 4H), 1.67 (d, J= 14.3 Hz, 3H), 2.03 (d, J= 14.0 Hz, IH), 2.30 (q, J= 9.8, 8.5 Hz, 4H), 2.65 (s, IH), 3.13 (q, J= 5.8 Hz, 2H), 3.34 (t, J= 6.2 Hz, 2H), 3.39 3.54 (m, 46H), 3.57 (t, J= 7.3 Hz, 2H), 3.64 (t, J= 6.2 Hz, 2H), 3.86 (s, 2H), 4.18 (d, J= 14.6 Hz, 2H), 4.49 (d, J= 19.2 Hz, IH), 4.93 (d, J= 4.8 Hz, IH), 5.07 (s, IH), 5.43 (s, IH), 5.50 (s, IH), 5.62 (d, J= 41.1 Hz, IH), 6.11 (s, IH), 6.20 6.36 (m, IH), 6.87 (d, J= 7.5 Hz, IH), 6.98 (s, 2H), 7.16 (t, J= 7.8 Hz, IH), 7.23 (t, J= 9.0 Hz, 3H), 7.34 (d, J= 8.4 Hz, 3H), 7.43 (d, J= 8.4 Hz, IH), 7.97 (s, IH), 9.80 (s, IH).
Example 24: N-(3-((4-((2S,6aS,6bR, 7S,8aS,8bS, I OR llaR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl oxo-2,4,6a,6b,7,8,8a,8b,I la,12, 12a,12b-dodecahydro-1H- naphtho[2',I':4,5]indeno[1,2-d][ 1,3]dioxolyl)phenyl)thio)phenyl)(2,5-dioxo-2,5-dihydro-IH­ pyrrol-l-yl)propanamide In a 4 mL vial -dioxo-2,5-dihydro-IH-pyrrol- l-yl)propanoic acid was added (43.5mg, 0.26mmol), foll owed by HATU (148mg, 0.39mmol) ved in DMA (1.0mL), foll owed by N,NDiisopropylethyl amine neat (67ul, 0.39mmol). Then a solution of (2S,6aS,6bR, 7S,8aS,8bS,IOR l laR,12aS,12bS)- l (3-aminophenyl)thio)phenyl)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl-6a,6b,7,8,8a,8b,I la,12, 12a,12b-decahydro-1H- naphtho[2', ]indeno[ l,2-d][l,3]dioxol-4(2H)-one(80.83mg, ol)(80.83mg, 0.13mmol) dissolved in DMA(0.5mL) was added. The reaction was shaken at room ature for 2 hours. The reaction was checked by LC/MS and purified by reverse phase HPLC (Method q, linear gradient 45 - 75%), to provide the title compound. LCMS (Method s, Table 7) Rt= 0.78 min; MS m/z =775.3 (M+H)\ 1H NMR (400 MHz, DMSO-d6/D20, Temp= 27 °C) 8 7.61 - 7.57 (m, lH), 7.49 - 7.44 (m, lH), 7.43 - 7.37 (m, 2H), 7.34 - 7.22 (m, 4H), 7.06 - 7.02 (m, lH), 6.92 (s, 2H), 6.29 (dd, J= 10.2, 1.9 Hz, lH), 6.14 - 6.09 (m, lH), 5.72 - 5.52 (m, lH), 5.46 (s, lH), 4.98 - 4.93 (m, lH), 4.52 (d, J= 19.4 Hz, lH), 4.26 - 4.14 (m, 2H), 3.73 - 3.71 (m, 2H), 3.69 - 3.65 (m, 2H), 2.73 - 2.55 (m, lH), 2.35 - 2.26 (m, lH), 2.25 - 2.12 (m, lH), 2.03 - 1.95 (m, lH), 1.79 - 1.62 (m, 3H), 1.55 - 1.39 (m, 4H), 0.85 (s, 3H).
Example 25: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,1 OR llaR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl oxo-2,4,6a,6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[1,2-d] [ oxol- l0-yl)phenyl)thio)phenyl)(2,5-dioxo-2,5-dihydro-lH­ pyrrol- l-yl)hexanamide Prepared as described in example 24 from 6-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l-yl)hexanoic acid. Purified by reverse phase HPLC (Metho ds, linear gradient 50 - 80%). LCMS d c, Table 7) Rt 0.82 min; MS m/z 817.3 (M+H)\ 1H NMR (400 MHz, DMSO-d6/D 27 °C) 8 7.68 - , Temp 7.65 (m, lH), 7.53 - 7.49 (m, lH), 7.42 - 7.38 (m, 2H), 7.33 -7.24 (m, 4H), 7.04 -7.01 (m, lH), 6.91 (s, 2H), 6.29 (dd, J 10.1, 1.9 Hz, lH), 6.14 -6.10 (m, lH), 5.72 - 5.53 (m, lH), 5.46 (s, lH), 4.96 -4.92 (m, lH), 4.51 (d, J= 19.4 Hz, lH), 4.24 - 4.15 (m, 2H), 3.38 (t, J= 7.0 Hz, 2H), 2.70 - 2.54 (m, lH), 2.35 - 2.11 (m, 5H), 2.03 -1.96 (m, lH), 1.76 -1.61 (m, 3H), 1.59 -1.41 (m, 8H), 1.24 -1.13 (m, 2H), 0.85 (s, Example 26: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS, 1 OR llaR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl-4 -oxo-2,4,6a,6b,7,8,8a,8b, lla, 12, 12a,12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[1,2-d] [ oxol- l0-yl)phenyl)thio)phenyl)(2,5-dioxo-2,5-dihydro-lH­ pyrrol- l-yl)benzamide Prepared as described in example 24 from 4-(2,5-dioxo-2,5-dihydro- rol- l-yl)benzoic acid. Purified by reverse phase HPLC (Method s, linear gradient 50 - 80%). LCMS (Metho d c, Table 7) Rt 0.83 min; MS m/z 823.2 (M+H)\ 1H NMR (400 MHz, DMSO-d6/D 27 °C) 8 8.02 - , Temp 7.97 (m, 2H), 7.88 - 7.85 (m, lH), 7.77 - 7.73 (m, lH), 7.52 -7.47 (m, 2H), 7.44 - 7.36 (m, 3H), 7.35 - 7.30 (m, 2H), 7.29 -7.23 (m, lH), 7.15 (s, 2H), 7.14 - 7.10 (m, lH), 6.29 (dd, J= 10.2, 1.9 Hz, lH), 6.15 - 6.09 (m, lH), 5.71 - 5.54 (m, lH), 5.47 (s, lH), 4.97 -4.94 (m, lH), 4.52 (d, J 19.4 Hz, lH), 4.24 -4.14 (m, 2H), 2.70 - 2.57 (m, lH), 2.37 - 2.27 (m, lH), 2.24 - 2.12 (m, lH), 2.03 -1.97 (m, lH), 1.75 -1.64 (m, 3H), 1.54 -1.42 (m, 4H), 0.85 (s, 3H).
Example 27: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,1 OR, llaR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl oxo-2,4,6a,6b,7,8,8a,8b,lla,12, 12a,12b-dodecahydro-1H- naphtho[2',l':4,5]indeno[ 1,2-d] [ l ,3]dioxol-l0-yl)phenyl)thio)phenyl)((2,5-dioxo-2,5-dihydro-lH­ pyrrol- l-yl)methyl)cyclohexanecarboxamide Prepared as described in example 24 from 4-((2,5-dioxo-2,5-dihydro-lH-pyrrol-lyl )methyl)cyclohexane-l-carboxylic acid. ed by reverse phase HPLC (Method q, linear gradient 50 -80%). LCMS (Method s, Table 7) R1= 0.85 min; MS m/z= 843.3 (M+H)\ 1H NMR (400 MHz, DMSO-d6/D20, Temp= 27 °C) 8 7.68 (t, J= 2.0 Hz, lH), 7.54 - 7.49 (m, lH), 7.42 -7.37 (m, 2H), 7.35 - 7.22 (m, 4H), 7.04 -7.01 (m, lH), 6.95 (s, 2H), 6.29 (dd, J= 10.1, 1.9 Hz, lH), 6.12 (s, lH), 5.71 - 5.53 (m, lH), 5.46 (s, lH), 4.99 -4.93 (m, lH), 4.51 (d, J= 19.4 Hz, lH), 4.25 - 4.15 (m, 2H), 3.26 (d, J= 7.0 Hz, 2H), 2.73 - 2.58 (m, lH), 2.35 -2.14 (m, 3H), 2.03 -1.96 (m, lH), 1.83 - 1.62 (m, 7H), 1.59 -1.40 (m, 5H), 1.37 -1.24 (m, 2H), 0.98 - 0.87 (m, 2H), 0.85 (s, 3H).
Example 28: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,1 OR, llaR,12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl oxo-2,4,6a,6b,7,8,8a,8b,lla,12, 12a,12b-dodecahydro-1H- o[2',l':4,5]indeno[l,2-d] [l,3]dioxol-l0-yl)phenyl)thio)phenyl)(3-(2,5-dioxo-2,5-dihydro-lH­ pyrrol-l-yl)propanamido)-3,6,9,12-tetraoxapentadecanamide 4] Prepared as described in example 24 from l-(2,5-dioxo-2,5-dihydro-lH-pyrrol- l-yl)oxo- 7, 10,13,16-tetraoxaazanonadecan- l9-oic acid. Purified by reverse phase HPLC d s, linear gradient 45 -75%). LCMS (Method c, Table 7)Rt 0.76 min; MS m/z 1022.4 (M+H)\ 1H NMR (400 MHz, DMSO-d6/D20, Temp 27 °C) 8 7.70 -7.66 (m, lH), 7.55 - 7.50 (m, lH), 7.43 - 7.37 (m, 2H), 7.34 -7.23 (m, 4H), 7.06 -7.01 (m, lH), 6.92 (s, 2H), 6.29 (dd, J 10.2, 1.9 Hz, lH), 6.14 - 6.11 (m, lH), .72 -5.53 (m, lH), 5.46 (s, lH), 5.00 -4.92 (m, lH), 4.51 (d, J 19.4 Hz, lH), 4.26 -4.15 (m, 2H), 3.66 (t, J 6.1 Hz, 2H), 3.59 (t, J 7.2 Hz, 2H), 3.51 - 3.40 (m, l lH), 3.33 (t, J 5.8 Hz, 2H), 3.12 (t, J 5.8 Hz, 2H), 2.70 - 2.58 (m, lH), 2.51 - 2.47 (m, 3H), 2.36 - 2.25 (m, 3H), 2.24 - 2.13 (m, lH), 2.04 - 1.97 (m, lH), 1.75 -1.65 (m, 3H), 1.56 - 1.42 (m, 4H), 0.85 (s, 3H).
Example 29: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,1 OR llaR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimeth yloxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol-l0-yl)phenyl)thio)phenyl)(2,5-dioxo-2,5-dihydro-lH­ pyrrol- l-yl)-3,6,9,12-tetraoxapentadecanamide tt,..___,,,O o--./" --./" ,..___,,, o o � Prepared as described in example 24 from l-(2,5-dioxo-2,5-dihydro-lH-pyrrol- l-yl)-3,6,9,12- tetraoxapentadecanoic acid. Purified by reverse phase HPLC d s, linear gradient 45 -75%).
LCMS (Method c, Table 7)Rt 0.80 min; MS m/z 951.3 (M+H)\ 1HNMR (400 MHz, DMSO-d6/D Temp 27 °C) 8 7.69 -7.66 (m, lH), 7.54 -7.49 (m, lH), 7.42 - 7.37 (m, 2H), 7.35 - 7.24 (m, 4H), 7.06 - 7.01 (m, lH), 6.93 (s, 2H), 6.29 (dd, J 10.2, 1.9 Hz, lH), 6.17 - 6.10 (m, lH), 5.71 - 5.55 (m, lH), 5.46 (s, lH), 4.98 -4.93 (m, lH), 4.51 (d, J 19.4 Hz, lH), 4.24 -4.16 (m, 2H), 3.66 (t, J 6.1 Hz, 2H), 3.56 - 3.51 (m, 2H), 3.50 -3.36 (m, 14H), 2.71 -2.60 (m, lH), 2.51 - 2.48 (m, 2H), 2.33 - 2.27 (m, lH), 2.18 (q, J 10.5 Hz, lH), 2.03 - 1.94 (m, lH), 1.74 -1.66 (m, 3H), 1.56 - 1.44 (m, 4H), 0.85 (s, 3H). e 30: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,1 OR l laR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, 12a, 12b-dodecahydro-1H- naphtho[2', l':4,5]indeno[ l,2-d][l,3]dioxol- l0-yl)phenyl)thio)phenyl)(2,5-dioxo-2,5-dihydro- lH­ pyrrol- 3,6,9,12,15, l8-hexaoxahenicosanamide "fflQ �o) ..,,,°'Jb 0 � '(.___,, 0--./'-o /'--./ O�0 Prepared as bed m example 24 from l-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l-yl)- 3,6,9,12,15,18-hexaoxahenicosanoic acid. Purified by reverse phase HPLC (Method q, linear gradient - 100%). LCMS (Method s, Table 7) R1 = 0.80 min; MS m/z did not ionize; 1H NMR (400 MHz, DMSO-d6/D20, Temp = 27 °C) 8 7.69 - 7.66 (m, lH), 7.55 - 7.50 (m, lH), 7.44 - 7.37 (m, 2H), 7.34 - 7.24 (m, 4H), 7.06 -7.01 (m, lH), 6.94 (s, 2H), 6.29 (dd, J = 10.2, 1.9 Hz, lH), 6.13 (s, lH), 5.69 -5.55 (m, lH), 5.46 (s, lH), 4.97 - 4.93 (m, lH), 4.51 (d, J = 19.4 Hz, lH), 4.23 -4.16 (m, 2H), 3.66 (t, J = 6.1 Hz, 2H), 3.56 - 3.38 (m, 22H), 2.70 - 2.63 (m, lH), 2.54 -2.53 (m, 2H), 2.51 -2.48 (m, 2H), 2.33 -2.26 (m, lH), 2.18 (q, J = 10.3 Hz, lH), 2.03 -1.97 (m, lH), 1.73 -1.65 (m, 3H), 1.55 -1.44 (m, 4H), 0.85 (s, Example 31: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,l0R,l laR 12aS, 12bS)-2,6b-difluorohydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, 12a, 12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[1,2-d][ l,3]dioxol- l0-yl)phenyl)thio)phenyl)(2-(2-(2-(2,5-dioxo-2,5-dihydrolH-pyrrol- l-yl)ethoxy)ethoxy)ethoxy)propanamide Prepared as described in example 24 from 3-(2-(2-(2-(2,5-dioxo-2,5-dihydro-lH-pyrrol- lyl )ethoxy)ethoxy )ethoxy )propanoic acid. Purified by reverse phase HPLC (Method q, linear gradient 45 - 75%). LCMS (Method s, Table 7) R1= 0.80 min; MS m/z= 908.1 (M+H)\ 1H NMR (400 MHz, DMSO­ d6/D2O, Temp= 27 °C) 8 7.69 -7.66 (m, lH), 7.54 -7.51 (m, lH), 7.42 -7.38 (m, 2H), 7.34 -7.24 (m, 4H), 7.06 -7.02 (m, lH), 6.93 (s, 2H), 6.29 (dd, J 10.2, 1.9 Hz, lH), 6.12 (s, lH), 5.68 -5.55 (m, lH), .46 (s, lH), 4.98 -4.94 (m, lH), 4.51 (d, J= 19.4 Hz, lH), 4.24 -4.16 (m, 2H), 3.64 (t, J= 6.1 Hz, 2H), 3.55 -3.50 (m, 2H), 3.47 -3.37 (m, 9H), 2.69 -2.66 (m, lH), 2.54 -2.53 (m, lH), 2.50 -2.47 (m, 2H), 2.32 -2.25 (m, lH), 2.21 -2.14 (m, lH), 2.03 -1.97 (m, lH), 1.74 -1.65 (m, 3H), 1.54 -1.43 (m, 4H), 0.85 (s, 3H).
Example 32: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,1 OR aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl oxo-2,4,6a,6b,7,8,8a,8b, l la,12, 12a, decahydro-1H- naphtho[2', l': 4,5]indeno[1,2-d][ l ,3]dioxol- l0-yl)phenyl)thio)phenyl)(2-(2-(3-(2,5-dioxo-2,5-dihydrolH-pyrrol-l-yl )propanamido )ethoxy)ethoxy)propanamide Prepared as described in example 24 from 3-(2-(2-(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol-lyl )propanamido)ethoxy)ethoxy noic acid. Purified by e phase HPLC (Method q, linear gradient 45 -75%). LCMS (Methods, Table 7) Rt 0.76 min; MS m/z 934.4 (M+H)\ 1H NMR (400 MHz, DMSO-d6/D20, Temp 27 °C) 8 7.69 -7.66 (m, lH), 7.54 - 7.49 (m, lH), 7.42 - 7.38 (m, 2H), 7.34 -7.23 (m, 4H), 7.05 -7.01 (m, lH), 6.91 (s, 2H), 6.29 (dd,J 10.2, 1.9 Hz, lH), 6.13 (s, lH), 5.68 - .56 (m, lH), 5.46 (s, lH), 4.97 -4.93 (m, lH), 4.51 (d, J 19.4 Hz, lH), 4.24 - 4.15 (m, 2H), 3.66 (t, J 6.2 Hz, 2H), 3.58 (t, J 7.2 Hz, 2H), 3.51 -3.43 (m, 4H), 3.33 (t, J 5.8 Hz, 2H), 3.10 (t, J 5.7 Hz, 2H), 2.63-2.58(m,1H), 2.55 -2.53 (m, lH), 2.50 - 2.49 (m, 2H), 2.32 -2.27 (m, 2H), 2.18 (q,J 10.3 Hz, lH), 2.03 -1.97 (m, lH), 1.73 -1.63 (m, 3H), 1.54 -1.42 (m, 4H), 0.85 (s, 3H).
Example 33: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,1 OR llaR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la,12, 12a, decahydro-1H- naphtho[2', l indeno[1,2-d] [ l ,3]dioxol- l0-yl)phenyl)thio)phenyl)(2,5-dioxo-2,5-dihydro- lH­ pyrrol- l-yl)acetamide o • i:qO .., A O"'" F HN O ��-··l �0 CH3 \lAsJv HO Prepared as described in example 24 from -dioxo-2,5-dihydro-lH-pyrrol- l-yl)acetic acid. Purified by reverse phase HPLC (Method s, linear gradient 45 - 75%). LCMS (Method c, Table 7) Rt 0.95 min; MS m/z 761.7 (M+H)\ 1H NMR (400 MHz, DMSO-d6/D 27 °C) 8 7.65 - , Temp 7.61 (m, lH), 7.52 -7.47 (m, lH), 7.44 - 7.39 (m, 2H), 7.36 (t,J 7.9 Hz, lH), 7.33 - 7.25 (m, 3H), 7.11 - 7.08 (m, lH), 7.07 (s, 2H), 6.32 (dd,J 10.1, 1.9 Hz, lH), 6.15 (s, lH), 5.72 - 5.55 (m, lH), 5.47 (s, lH), .00 -4.92 (m, lH), 4.53 (d,J 19.5 Hz, lH), 4.30 -4.17 (m, 4H), 2.72 - 2.61 (m, lH), 2.38 -2.28 (m, lH), 2.19 (q,J 10.3 Hz, lH), 2.05 -1.98 (m, lH), 1.78 -1.64 (m, 3H), 1.60 -1.42 (m, 4H), 0.87 (s, 3H).
Example 34: N-(3-((4-((2S,6aS,6bR,7S,8aS,8bS,l0R,llaR12aS,12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la,12, 12a, 12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[1,2-d] [1,3]dioxolyl)phenyl)thio)phenyl)(2-(2-(2,5-dioxo-2,5-dihydro-lH­ pyrrol- l-yl)ethoxy )ethoxy )propanamide Prepared as described in e 24 from 3-(2-(2-(2,5-dioxo-2,5-dihydro-lH-pyrrol- lyl y)ethoxy)propanoic acid. Purified by reverse phaseHPLC (Method q, linear gradient 40 - 75%).
LCMS d c, Table 7) R1 0.95 min; MS m/z 863.9 (M+H)\ 1HNMR (400 MHz, DMSO-d6/D 20, Temp 27 °C)8 7.71 - 7.66 (m, lH), 7.55 - 7.51 (m, lH), 7.44 - 7.38 (m, 2H), 7.36 - 7.25 (m, 4H), 7.08 - 7.03 (m, lH), 6.91 (s, 2H), 6.31 (dd, J 10.1, 1.9 Hz, lH), 6.14 (s, lH), 5.72 - 5.55 (m, lH), 5.47 (s, lH), 4.98 -4.94 (m, lH), 4.53 (d, J= 19.4 Hz, lH), 4.27 - 4.14 (m, 2H), 3.63 (t, J= 6.1 Hz, 2H), 3.53 -3.43 (m, 8H), 2.72 - 2.61 (m, lH), 2.48 (t, J 6.2 Hz, 2H), 2.35 - 2.24 (m, lH), 2.20 (q, J 10.4 Hz, lH), 2.05 - 1.96 (m, lH), 1.76 - 1.65 (m, 3H), 1.57 - 1.41 (m, 4H), 0.87 (s, 3H).
Example 34A: Synthesis of 3-(2,5-dioxo-2,5-dihydro-lH-pyrrol- l-yl)-N-((S)-l-(((S)-l-((3-(4- ((6aR6bS,7S,8aS,8bS, lOR, l laR,12aS,12bS)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b, l 12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l 0- yl)benzyl)phenyl)amino)oxopropanyl)amino)oxopropanyl)propanamide 1] Step 1: Synthesis of (S)amino-N-((S)((3-(4- ((6aR6bS,7S,8aS,8bS, lOR, l laR,12aS,12bS)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l 0- yl)benzyl)phenyl)amino)oxopropanyl)propanamide 1) Fmoc Ala-Ala-OH HATU, 2,6-lut. 2) El2NH, 3h HATU (601 mg, 1.580 mmol) and 2,6-lutidine (0.37 mL, 3.16 mmol) were added to a 0 °C solution of (S)( (S)((((9H-fluorenyl)methoxy)carbonyl)amino)propanamido)propanoic acid (765 mg, 2.00 mmol), (6aR6bS,7S,8aS,8bS, l0R l laR12aS,12bS)(4-(3-aminobenzyl)phenyl)hydroxy- 8b-(2-hydroxyacetyl)-6a,8a-dimethyl-6a,6b,7,8,8a,8b, l la,12,12a,12b-decahydro-1H- naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-4(2H)-one (600 mg, 1.053 mmol) in DCM (6 mL) and DMF (12 mL). After 30 min, the mixture was warmed to room temperature and stirred overnight. Diethylamine (2.18 mL, 21.06 mmol) was added to the reaction mixture, and stirring ued at room temp for 3 h, whereupon volatile solvents were removed under reduced pressure. The residue was dissolved in 1: 1 DMSO:MeOH (12 mL) and purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 50 mm column). A gradient ofMeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of 90 mL/min (0-5.0 min 15% A, 5.0-20 min linear gradient 15-85% A, hold 5 min). Combined product fractions were lyophilized to give the title compound as an off-white solid (447 mg, 0.628 mmol, 60% yield). LC-MS (Method r, Table 7) Rt 0.78 min, m/z 711.9 [M+H]. 1H NMR (501 MHz, DMSO-d .03 (s, lH), 8.63 (d, J 7.2 Hz, lH), 8.07 (d, J 5.4 Hz, 3H), 7.44 7.38 (m, 2H), 7.38 7.34 (m, 2H), 7.29 (d, J 10.1 Hz, lH), 7.23 7.16 (m, 3H), 6.90 (dt, J 7.7, 1.3 Hz, lH), 6.14 (dd, J 10.1, 1.9 Hz, lH), 5.90 (t, J 1.6 Hz, lH), 5.38 (s, lH), 4.90 (d, J 5.3 Hz, lH), 4.52 4.37 (m, 2H), 4.27 (q, J 3.3 Hz, lH), 4.16 (d, J= 19.4 Hz, lH), 3.87 (s, 2H), 2.58 2.49 (m, lH), 2.28 (ddd, J= 13.4, 4.5, 2.1 Hz, lH), 2.09 (<ltd, J 17.0, 10.6, 5.0 Hz, lH), 2.00 (dd, J 12.2, 5.7 Hz, lH), 1.78 1.54 (m, 5H), 1.37 (s, 3H), 1.35 (s, 3H), 1.30 (d, J 7.1 Hz, 3H), 1.01 (ddd, J 22.1, 11.9, 4.2 Hz, 2H), 0.84 (s, 3H).
Step 2: sis of 3-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l-yl)-N-((S)- )((3-(4- ((6aR6bS,7S,8aS,8bS,1OR,l laR, 12aS, 12bS)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo- 2,4,6a,6b, 7,8,8a,8b, l la, 12, 12a, 12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol-l 0- yl)benzyl)phenyl)amino)oxopropanyl)amino)oxopropanyl)propanamide N,N-Diisopropylethylamine (0.33 mL, 1.875 mmol) was added to a room temperature solution ofN-succinimidyl 3-maleimidopropionate (250 mg, 0.938 mmol) and amino-N-((S)((3- ( R,6bS,7S,8aS,8bS,1 OR l laR12aS,12bS)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo- 2,4,6a,6b, 7,8,8a,8b, l la, 12, 12a, 12b-dodecahydro-1H-naphtho[2',l ':4,5]indeno[l,2-d][l,3]dioxol-l 0- yl)benzyl)phenyl)amino)oxopropanyl)propanamide (445 mg, 0.625 mmol) in DMF (12 mL). After min at room ature, the volatile solvents were removed under reduced pressure. The residue was d with 1: 1 DMSO:MeOH (12 mL) and purified by reverse phase HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 50 mm column). A gradient ofMeCN (A) and 0.1 % TFA in water (B) was used, at a flow rate of 90 mL/min (0-5.0 min 25% A, 5.0-20 min linear gradient 25-90% A, hold 5 min).
Combined product fractions were lyophilized to give the title compound as an off-white solid (295 .1 mg, 0.342 mmol, 55% yield). LC-MS (Method r, Table 7) Rt 0.85 min, m/z 863.4 [M+H]. 1H NMR (501 MHz, DMSO-d6) 8 9.71 (s, lH), 8.17 (d, J= 7.0 Hz, lH), 8.03 (d, J= 7.3 Hz, lH), 7.43 (dd, J= 7.8, 1.1 Hz, 2H), 7.38 7.32 (m, 2H), 7.29 (d, J 10.1 Hz, lH), 7.22 7.15 (m, 3H), 6.96 (s, 2H), 6.88 (dt, J 7.8, 1.3 Hz, lH), 6.13 (dd, J 10.1, 1.9 Hz, lH), 5.90 (t, J 1.6 Hz, lH), 5.37 (s, lH), 4.90 (d, J 5.4 Hz, lH), 4.48 (d, J= 19.4 Hz, lH), 4.32 (p, J = 7.1 Hz, lH), 4.27 (q, J = 3.3 Hz, lH), 4.21 (p, J = 7.1 Hz, lH), 4.16 (d, J= 19.4 Hz, lH), 3.87 (s, 2H), 3.59 (t, J= 7.3 Hz, 2H), 2.57 2.49 (m, lH), 2.38 (dd, J= 8.0, 6.6 Hz, 2H), 2.32 2.24 (m, lH), 2.15 2.04 (m, lH), 2.04 1.95 (m, lH), 1.80 1.54 (m, 5H), 1.37 (s, 3H), 1.26 (d, J = 7.1 Hz, 3H), 1. 15 (d, J= 7.1 Hz, 3H), 1.02 (ddd, J= 21.2, 12.1, 4.2 Hz, 2H), 0.84 (s, 3H).
Example 35: The following compounds were prepared using the methods described above.
Cpd. No. ure -··•07"'0- .,,o hQ 0 NH i= o J)--i-fo N H 0 ... ,0.,,o 7 ... ,o-hQ 0 y.'('­ 72 HOh Ht( h 0 o '-(° o -- \__ 75 o O o J-- N,,.____)l l �J � � fr( OH o O = H 76 0 78 r J-,,o �,�i)- ,QO ;OH 0 H O , H 79 0--1� J- ,J, 1 UL 0 4 � , N�O OH H,., nO , H '°o/J•1 0 '<'� :�□")r \_ N> 1 "'Jr H 0 0 N J 0 = H-ll� ,:;-- O Ov"" A I '>-. ,, 0 '•( ',.o,,, �F O . H ' 84 OMe O'.
H O � H OH NJ N � ...L,; O , N �, o II 0 a H\ 85 ))" ' }- ,J, l :J r(Y' OH ¼ , N� H/r( = H 86 ,JL) "r:, }- ,J, l :J � OH ¼ , Nu 0 = H 87 (V' ,JO }- ,J, I :J OH ¼ = , N� H/r( H 100 J- .,J , I I i ri, r,, 'C , S 0 4 (" N� ) H'n ' 0 = H F 101 0"'( � Jl .J � r"') �,,. n y-" I I HO 0 ½ N O = H 104 q �J � I / � ,,,. J:.�: o JN l , N . /2 H,,. n - H 0 � OH 0 0 J -- ro , U� � l/� Dv 0 105 HO 109 o �J .Y')HO 0,, ..lo J-- N�N l N�OH H/r( , �o O = H 110 o O �J () r-7) ,,..l_ o, ¼ H/r( : N�S��N l)l N O = H H o•· 112 6 I �J Y) 0,,..
J-- N�N , N OH o O = H 114 0 O o•· f H H 0 s:Lg"!'�-\(usd- OH ¼J,.� N I �"N°' S '""' H/")' = H , OH 115 0 0 ,.AN H J 07 ('o O rf'"Y 0Y") 116 ) ( O Cofo ( O ( 0------._)l_ N� (O H �J-- o--.,) l___,,, o o--.,) H o 0 O HO 0 0)fO fnl____,,o � O V v,,,.(O H 117 H �N 0 '-,,,"""- II O HO 0 0 0 121 O' 0 0 0 0,,•· ¼ OH }-N�N�SH 0 _, -S l__, N s rir o -o, (0 " 127 0 �o, 0 (' o rn � S 0 O) l_ o o 'Q' Y) o 128 Q: v,,h 9-'�Ct___ti!'�O''O,.<o " 129 0 O 0 l(N�o ('yNYYsYil o l___,,o o V V,,"(o H 131 0 LC-MSData ata 1H NMR (DMSO-d 6) 8: 0.79-0.87 (m, l0H), 0.95-1.13 (m, lH), 1.29 (d,J 7.lHz, 3H), 1.38 (s, 3H), 1.57-1.76 (m, 2H), 1.76 (s, 2H), 1.90-2.01 (m, Method a LC- 2H), 2.05 (s, 2H), 2.28 (s, 2H), 2.43 (dd,J 14.4, 7.0Hz, 2H), 3.58 (p,J MS, Table 7 6.8Hz, 2H), 4.11 (dd,J 8.4, 6.7Hz, lH), 4.16 (d,J 19.4Hz, lH), 4.28 Rt 1.28 min; 70 (s, lH), 4.36 (q,J 6.7Hz, lH), 4.49 (d,J 19.4Hz, lH), 4.75 (s, lH), m/z 894.0 4.90 (d,J 5.lHz, lH), 5.40 (s, lH), 5.90 (s, lH), 6.14 (dd,J 10.1, [M+H+] l.9Hz, lH), 6.89-7.02 (m, 6H), 7.29 (d,J 10.lHz, lH), 7.42 (d,J 8.7Hz, 2H), 7.56-7.63 (m, 2H), 7.98 (d,J 8.4Hz, lH), 8.13 (d,J 6.9Hz, lH), 9.86 (s, lH) 1H NMR (DMSO-d 6) 8: 0.85 (s, 3H), 1.16 (d,J 7.lHz, 3H), 1.29 (d,J Method r, 7.lHz, 3H), 1.48 (s, 3H), 1.53 (d,J 12.6Hz, lH), 1.60-1.77 (m, 3H), Table 7 1.98-2.09 (m, lH), 2.24 (dd,J 21.2, 8.6Hz, 2H), 2.39 (dd,J 8.0, 6.5Hz, 2H), 2.53-2.72 (m, lH), 3.59 (dd,J 8.1, 6.5Hz, 2H), 4.13-4.27 Rt 0.79 min; (m, 2H), 4.34 (p,J 7.lHz, lH), 4.51 (dd,J 19.5, 6.4Hz, lH), 4.93 (d, m/z 900.91 J 5.0Hz, lH), 5.07 (t,J 6.0Hz, lH), 5.45 (s, lH), 5.50 (dd,J 4.5, [M+H+] 1.7Hz, lH), 5.53-5.75 (m, lH), 6.07-6.12 (m, lH), 6.27 (dd,J 10.2, l.9Hz, lH), .00 (m, 6H), 7.24 (dd,J 10.2, l.4Hz, lH), 7.36-7.43 (m, 2H), 7.58-7.66 (m, 2H), 8.08 (d,J 7.3Hz, lH), 8.18 (d,J 7.0Hz, lH), 9.83 (s, lH) 1HNMR(MeOH-d 12.2, 7.1 Hz, 3H), 1.48 Method m 4) 8: 1.00 (s, 3H), 1.37 (dd,J (t,J 7.2 Hz, 3H), 1.59 (s, 4H), 1.69 (dd,J 27.0, 13.1 Hz, 2H), 1.79 LC-MS, (dd,J 13.7, 5.8 Hz, 2H), 2.26 (d,J 13.6 Hz, lH), 2.38 (d,J 8.0 Hz, Table 7 3H), 2.56 (td,J 12.5, 11.2, 6.8 Hz, 3H), 2.60 - 2.81 (m, lH), 3.80 (dt,J Rt 1.71 min; 72 12.7, 6.8 Hz, 2H), 4.24 (dd,J 11.9, 7.0 Hz, lH), 4.32 (s, 2H), 4.43 - m/z 917 + 4.51 (m, lH), 4.64 (d,J 19.4 Hz, lH), 5.07 (d,J 4.6 Hz, lH), 5.47 (s, [M+H ] lH), 5.57 (d,J 42.9 Hz, lH), 6.27- 6.38 (m, 3H), 6.73 (d,J 3.0 Hz, 2H), 7.16- 7.25 (m, 2H), 7.36 (dt,J 16.7, 8.0 Hz, 6H), 7.70 (dd,J 22.7, 8.4 Hz, 2H) Method m 1HNMR(MeOH-d LC-MS, 4) 8: 1.00 (s, 3H), 1.49 (d,J= 7.2 Hz, 3H), 1.59 (s, 3H), 1.60 - 1.89 (m, 3H), 2.04 (d,J 52.2 Hz, lH), 2.27 (d,J 13.5 Hz, Table 7 lH), 2.31-2.52 (m, 4H), 2.58 (t,J 6.7 Hz, 2H), 3.81 (t,J 6.7 Hz, 72 2H), 4.34 (d,J 19.7 Hz, 3H), 4.42 - 4.53 (m, lH), 4.64 (d,J 19.4 Hz, Rt 1.88 min; lH), 5.07 (d,J 4.6 Hz, lH), 5.47 (s, lH), 5.58 (d,J 40.9 Hz, lH), 6.25 m/z 975 + - 6.47 (m, 2H), 6.76 (s, 2H), 7.20 (d,J 8.1 Hz, 2H), 7.28- 7.44 (m, [M+H ] 5H), 7.67 (d,J 8.5 Hz, 2H) 1HNMR(DMSO-d 7.1 Hz, 3H), 1.27 (d,J 6) 8: 0.84 (s, 3H), 1.16 (d,J 7.1 Hz, 3H), 1.48 (s, 4H), 1.59- 1.77 (m, 3H), 1.96- 2.08 (m, lH), 2.13 Method a LC- - 2.33 (m, 2H), 2.39 (dd,J 7.9, 6.7 Hz, 2H), 2.52 (s, lH), 2.53- 2.72 MS, Table 7 (m, lH), 3.59 (t,J 7.3 Hz, 2H), 4.08 -4.26 (m, 5H), 4.32 (p,J 7.0 Hz, Rt 2.08 min; 73 lH), 4.50 (d,J 19.4 Hz, lH), 4.93 (d,J 5.0 Hz, lH), 5.44 (s, lH), 5.45 m/z 931.30 - 5.51 (m, lH), 5.63 (dt,J 48.4, 9.3 Hz, lH), 6.11 (d,J 2.1 Hz, lH), [M+H+] 6.28 (dd,J 10.2, 1.9 Hz, lH), 6.93- 7.02 (m, 3H), 7.19 (t,J 8.0 Hz, lH), 7.24 (dd,J 10.1, 1.4 Hz, lH), 7.30- 7.44 (m, 5H), 7.65 (t,J 1.9 Hz, lH), 8.05 (d,J 7.2 Hz, lH), 8.16 (d,J 7.0 Hz, lH), 9.77 (s, lH) 1HNMR(DMSO-d 7.lHz, 3H), 1.26 (d,J 6) 8: 0.84 (s, 3H), 1.15 (d,J Method r, 7.2Hz, 4H), 1.48 (s, 4H), 1.59-1.79 (m, 3H), 1.94-2.10 (m, lH), 2.10-2.31 Table 7 (m, 2H), 2.37 (t,J 7.3Hz, 2H), 2.51-2.77 (m, lH), 3.58 (t,J 7.3Hz, 2H), 4.10-4.25 (m, 3H), 4.31 (p,J 7.lHz, lH), 4.51 (d,J 19.4Hz, lH), 74 Rt 0.82 min; 4.94 (d,J 5.0Hz, lH), 5.45 (s, lH), 5.50 (s, lH), 5.62 (dt,J 48.6, m/z 918.60 9.4Hz, lH), 6.10 (s, lH), 6.27 (dd,J 10.1, l.9Hz, lH), 6.96 (s, 2H), [M+H ] 7.02 (dd,J 7.3, 1.7Hz, lH), 7.20-7.34 (m, 4H), 7.40 (d,J 8.3Hz, 2H), 7.58 (dd,J 7.9, 2. lHz, lH), 7.69 (d,J 2.lHz, lH), 8.07 (d,J 7.2Hz, lH), 8.16 (d,J 7.0Hz, lH), 9.89 (s, lH) 1HNMR(DMSO-d 7.1 Hz, 3H), 1.24 (d,J 6) 8: 0.83 (s, 3H), 1.13 (d,J 7.2 Hz, 3H), 1.46 (s, 4H), 1.57- 1.77 (m, 3H), 2.01 (dt,J 13.9, 3.7 Hz, Method r, lH), 2.13- 2.32 (m, 2H), 2.36 (dd,J 8.0, 6.7 Hz, 2H), 2.51- 2.73 (m, Table 7 lH), 3.56 (d,J 7.3 Hz, 2H), 3.85 (s, 2H), 4.10- 4.25 (m, 3H), 4.30 (p,J 7.1 Hz, lH), 4.47 (d,J 19.4 Hz, lH), 4.91 (d,J 4.9 Hz, lH), 5.41 (s, Rt 0.82 min; lH), 5.48 (s, lH), 5.51- 5.71 (m, lH), 6.09 (d,J 2.0 Hz, lH), 6.26 (dd, m/z 899.87 J 10.2, 1.9 Hz, lH), 6.87 (dt, J 7.6, 1.3 Hz, lH), 6.95 (s, 2H), 7.16 (t, [M+H+] J 7.8 Hz, lH), 7.18- 7.27 (m, 3H), 7.32 (d,J 8.1 Hz, 2H), 7.39 (d,J 1.9 Hz, lH), 7.43 (dd, J= 8.5, 1.9 Hz, lH), 8.01 (d,J= 7.2 Hz, lH), 8.14 (d, J= 7.0 Hz, lH), 9.70 (s, lH) 1HNMR(DMSO-d 7.4 Hz, 3H), 1.19 (dt,J 33.9, 7.1 Method r, 6) 8: 0.88 (d,J Table 7 Hz, llH), 1.35- 1.63 (m, l0H), 1.61- 1.85 (m, 2H), 2.06 (q,J 7.4 Hz, 3H), 2.16 - 2.35 (m, lH), 2.38 (t,J 7.3 Hz, lH), 3.87 (d,J 8.1 Hz, 76 2H), .42 (m, 3H), 4.67- 5.15 (m, 2H), 5.51- 5.73 (m, 3H), 6.11 Rt 1.00 min; (s, lH), 6.28 (dd,J 10.1, 2.1 Hz, lH), 6.89 (d, J 7.3 Hz, lH), 6.92- m/z not 7.02 (m, 3H), 7.13- 7.29 (m, 5H), 7.30- 7.41 (m, 2H), 7.42- 7.57 (m, observed lH), 7.88- 8.34 (m, 2H), 9.74 (s, lH) 1H NMR (DMSO-d 6) 8: ,3H),1.26(dd, J .5 Hz, 9H),1.50 (s,4H),1.70 (t, J 8.4 Hz,3H), 1.90 ,2H),2.14 2.35(m, lH), Methodm, 2.35 2.45(m,lH),2.54 2.77 (m,lH),2.96(ddd, J= 47.6, 15.3, 6.6 Table 7 Hz,2H), 3.57(t,J 7.3 Hz,2H), 3.89 (s, 2H), 4.20(d,J 19.0 Hz,lH), 4.32(dt, J 27.9, 7.0 Hz, 2H),4.45 4.64(m,2H),4.94(d,J 4.7 Hz, Rt 1.62 min; lH),5.07(d, J 39.6 Hz,lH),5.45(s, lH),5.49 5.79(m,2H),6.12(s, m/z 1058.3 lH),6.29 (dd, J 10.2,1.8 Hz,lH), 6.92(d,J 7.6 Hz,lH), 6.99(s, ] 2H),7.20 (t, J 7.9 Hz, 25(t,J 8.6 Hz,3H), 7.33 7.42(m, 4H),7.45(d, J 8.1 Hz,lH),8.31 (d,J 8.0 Hz,lH),8.95(s,lH),9.88 (s, lH),14.10(s,2H) 1H NMR d 6) 8: 0.84(s,3H),1.17 (d, J 7.1 Hz,3H), 1.25(d, J 7.1 Hz,3H),1.48(s,4H), 1.57(q, J 6.2 Hz,4H),1.68(dq, J 13.7,6.3, Method r, .6 Hz,3H), 1.99 2.06(m,lH),2.09 ,2H),2.18 , Table 7 2H),2.55 2.72(m,3H),2.78(s,4H),3.87(s, 2H),4.14 4.22 (m,2H), 4.26(p, J 7.1 Hz,lH),4.33(p,J 7.1 Hz,lH),4.49(d, J 19.4 Hz, 78 Rt 0.80 min; lH),4.93 (d, J 5.1 Hz,lH),5.43(s,lH),5.49(d, J 5.4 Hz,lH),5.54 m/z 1005.1 .75(m,lH),6.11(s, lH),6.28(dd,J 10.2, 2.0 Hz, lH), 6.89 (d, J [M+MeOH+H +] 7.6 Hz,lH),7.17(t, J 7.9 Hz,lH),7.23 (t, J 9.7 Hz, 3H), 7.34 (d, J 7.8 Hz,2H), 7.39(s,lH),7.44(d, J 8.1 Hz,lH),7.99(d,J 7.2 Hz, lH),8.02 (d, JJ 7.3 Hz,lH), 9.77 (s, lH) 1HNMR(DMSO-d 6) 8: 0.85(s,3H),1.14(d,J 7.1 Hz,3H), ,J 7.1 Hz,3H),1.48(s, 4H),1.61 1.80 (m,3H),2.04(d, J 13.1 Hz, lH), Method r, 2.25(ddd, J 18.6, 14.9,8.4 ,2.37 (dd, J 8.0,6.5 Hz,2H), 2.53 Table 7 2.74(m,lH),3.57(t, J 7.3 Hz,2H),4.09 4.24(m,2H), 4.30 (p,J 7.1 Hz,lH), 4.52(dd, J 19.5,6.4 Hz,lH),4.94(d,J 5.0 Hz,lH), Rt 0.80 min; 5.08(t, J 5.9 Hz,lH),5.46(s,lH),5.48 ,lH),5.63(dt, J m/z 901.81 48.9,9.1 Hz, lH),6.10(s, lH),6.27 (dd, J 10.1, 1.9 Hz, lH),6.69 (ddd, [M+H+] J 7.9,2.6,1.1 Hz, 96(s,2H),6.98 7.06 (m,2H),7.22 7.32 (m,2H),7.32 7.40(m,2H), 7.39 7.51(m,2H),8.06(d,J= 7.2 Hz, (d, J 7.0 Hz,lH),9.87 (s,lH) 1H NMR(MeOH-d 4) 8: 1.00(s, 3H), 1.36 (dd, J 11.5,7.1 Hz,4H), 1.46 (t, J 6.9 Hz,3H), 1.53 1.76 (m,5H),1.75 1.89 (m,2H),2.28(d,J Methodm, Table 7 13.8 Hz,lH), 2.33 2.48(m,lH),2.48 2.62(m,2H),2.61 2.84(m, lH),3.72 3.88 (m,2H),3.95(s,2H),4.18 4.40(m, 3H), 4.46 (q,J 80 6.9 Hz,lH),4.65(d, J= 19.4 Hz,lH),5.07(d, J= 4.6 Hz,lH),5.43 Rt 1.64 min; .69(m,2H), 6.30 6.39 (m, 2H),6.69(s,2H),7.14(dd, J= 8.2,5.6 Hz, m/z 899 2H),7.23 (dd, J 7.9,3.4 Hz,3H), 7.34(d,J 10.1 ,7.38(d,J [M+H+] 7.8 Hz,2H),7.51(d, J 8.2 Hz,2H),7.55(d, J 8.2 Hz,lH),8.55(s, 1H NMR(MeOH-d 4) 8: 0.88(s, 3H), 1.21(d, J 11.6 Hz,3H),1.35(d, J Methodm, 7.1 Hz,3H),1.48(s, 3H), 1.57(dd, J 25.8,13.5 Hz,2H),1.68(dd,J Table 7 13 .4, 5.5 Hz,2H),1.83(dd, J 14.4,7.5 Hz,lH),1.88 2.05(m,lH), 2.12 2.21 (m,lH),2.30 (q, J 12.7,10.3 Hz, 3H),2.44(t, J 6.7 Hz, 81 Rt 1.526 2H),2.49 2.73(m,lH),3.68(t, J 6.7 Hz,2H),3.82(s, 2H),4.11 mm; ,3H),4.34(q,J 7.1 Hz,lH), 4.53(d, J 19.4 Hz, lH),4.95(d, m/z 957 J 4.6 Hz,lH),5.30 5.59(m,2H),6.23 (dd, J 13.2, 3.0 Hz, 2H),6.60 [M+H+] (s,2H),7.02(d,J 8.2 Hz,2H), 7.10(d,J 7.9 Hz,2H),7.24(dd,J .1,8.9 Hz,3H),7.37(d,J 8.2 Hz,2H) 1H NMR (DMSO-d Method r, 6) 8: 0.84 (s, 3H), 0.96 (s, 2H), 1.01 1.19 (m, 4H), Table 7 1.19 1.35 (m, 2H), 1.48 (s, 3H), 1.67 (d, J 14.3 Hz, 2H), 2.03 (d, J 19.7 Hz, IH), 2.13 2.42 (m, 5H), 2.64 (d, J 8.4 Hz, 2H), 3.08 (s, 3H), 82 3.38 3.61 (m, IH), 3.93 (s, 2H), 4.17 (d, J 18.9 Hz, 3H), 4.49 (d, J Rt 0.77 min; 19.3 Hz, IH), 4.92 (d, J 4.8 Hz, IH), 5.43 (s, IH), 5.49 (s, IH), 5.53 913.27 .77 (m, IH), 6.11 (s, IH), 6.28 (dd, J 10.2, 1.8 Hz, IH), 6.96 (d, J [M+H+] 6.1 Hz, IH), 7.10 7.41 (m, 8H), 7.83 8.20 (m, IH) 1H NMR (DMSO-d 6) 8: 0.84 (s, 3H), 0.93 (dd, J 23.5, 6.9 Hz, 3H), 1.04 1.16 (m, 3H), 1.48 (s, 4H), 1.60 1.76 (m, 3H), 2.03 (d, J 20.7 Hz, Method r, IH), 2.17 2.26 (m, IH), 2.24 2.40 (m, 2H), 2.55 2.72 (m, 2H), 2.96 Table 7 (d, J 13.0 Hz, 3H), 3.55 (t, J 7.3 Hz, IH), 3.72 (d, J 57.6 Hz, 3H), 3.93 (d, J 4.7 Hz, 2H), 4.10 4.28 (m, 4H), 4.49 (d, J 19.5 Hz, IH), Rt 0.88 min; 4.93 (d, J 5.0 Hz, IH), 5.44 (d, J 2.9 Hz, IH), 5.50 (d, J 4.2 Hz, m/z 943.52 IH), 5.63 (dt, J 48.7, 9.8 Hz, IH), 6.11 (s, IH), 6.28 (dd, J 10.2, 1.9 [M+H+] Hz, IH), 6.82 (t, J 7.l Hz, IH), 6.93 7.00 (m, IH), 7.02 7.07 (m, IH), 7.19 (d, J 7.9 Hz, IH), 7.24 (d, J I0.0 Hz, IH), 7.29 7.41 (m, 4H), 7.93 (d, J 7. l Hz, IH) 1H NMR d 6) 8: 0.83 (s, 3H), 1.17 (d, J 7. l Hz, 3H), 1.25 (d, J 7.1 Hz, 3H), 1.46 (s, 4H), 1.58 1.77 (m, 3H), 2.01 (dt, J 13.8, 4.1 Hz, Method r, Table 7 IH), 2.23 (<ltd, J 25.2, 12.3, 10.9, 5.8 Hz, 2H), 2.31 2.40 (m, 2H), 2.49 2.73 (m, IH), 3.56 (t, J 7.3 Hz, 2H), 3.74 (s, 3H), 3.84 (s, 2H), 4.11 4.20 (m, 2H), 4.27 (p, J 7.l Hz, IH), 4.39 (p, J 7.2 Hz, IH), 84 Rt 0.82 min; 929.45 4.47 (d, J 19.4 Hz, IH), 4.91 (d, J 4.8 Hz, IH), 5.41 (s, IH), 5.42 .50 (m, IH), 5.50 5.78 (m, IH), 6.09 (s, IH), 6.26 (dd, J IO.I, 1.9 [M+H+] Hz, IH), 6.70 (dd, J 8.3, 1.8 Hz, IH), 6.89 (d, J 1.8 Hz, IH), 6.95 (s, 2H), 7.23 (d, J 8.5 Hz, 3H), 7.31 (d, J 7.9 Hz, 2H), 7.87 (d, J 8.2 Hz, IH), 8.14 (d, J 7.5 Hz, IH), 8.23 (d, J 7.2 Hz, IH), 8.81 (s, IH) 1H NMR (DMSO-d 6) 8: 0.84 (s, 3H), 1.16 (d, J 7. l Hz, 3H), 1.27 (d, J 7.1 Hz, 3H), 1.39 (qd, J 13.1, 5.2 Hz, IH), 1.48 (s, 3H), 1.55 1.72 (m, Method r, Table 7 3H), 1.78 1.90 (m, IH), 2.03 (d, J 25.4 Hz, IH), 2.13 (td, J 12.2, 6.8 Hz, IH), 2.29 2.41 (m, 3H), 2.48 (p, J 1.9 Hz, IH), 2.58 2.69 (m, IH), 3.59 (t, J 7.3 Hz, 2H), 4.09 4.26 (m, 3H), 4.29 4.44 (m, 3H), Rt 0.73 min; m/z 897.3 4.47 (d, J 19.4 Hz, IH), 4.82 4.91 (m, IH), 5.32 (s, IH), 5.41 (s, IH), 6.02 (d, J l.7 Hz, IH), 6.22 (dd, J IO. I, 1.9 Hz, IH), 6.47 (d, J 8.7 [M+H+] Hz, IH), 6.97 (s, 2H), 7.18 7.21 (m, 2H), 7.23 (t, J 6.2 Hz, IH), 7.27 (d, J IO.I Hz, IH), 7.33 (dd, J 8.7, 2.3 Hz, IH), 7.44 7.57 (m, 2H), 7.95 (d, J 2.3 Hz, IH), 8.04 (d, J 7.3 Hz, IH), 8.10 8.23 (m, 2H), 9.73 (s, IH) 1H NMR (DMSO-d 6) 8: 0.83 (s, 3H), 1. 15 (d, J 7.2 Hz, 3H), 1.26 (d, J 7.1 Hz, 3H), 1.36 (d, J 6.7 Hz, 4H), 1.48 (s, 3H), 1.63 (t, J I I. I Hz, Method r, 3H), 1.74 1.90 (m, IH), 1.93 2.19 (m, 2H), 2.26 2.41 (m, 3H), 2.48 Table 7 (p, J 1.8 Hz, IH), 2.58 2.70 (m, IH), 3.59 (t, J 7.3 Hz, 2H), 4.09 4.27 (m, 3H), 4.33 (p, J 7.2 Hz, IH), 4.46 (d, J 19.4 Hz, IH), 4.85 (d, Rt 0.73 min; J 4. l Hz, IH), 4.93 (t, J 6.9 Hz, IH), 5.04 (s, IH), 5.30 (s, IH), 5.40 m/z 911.46 (dd, J 4.5, 1.7 Hz, IH), 6.02 (s, IH), 6.21 (dd, J IO.I, 1.9 Hz, IH), [M+H+] 6.48 (s, IH), 6.97 (s, 2H), 7.26 (t, J 9.3 Hz, 3H), 7.32 (d, J 8.0 Hz, IH), 7.48 (d, J 8.3 Hz, 2H), 7.90 (d, J 2.2 Hz, IH), 8.03 (d, J 7.3 Hz, IH), 8.16 (d, J 7.0 Hz, IH), 9.71 (s, IH) 1HNMR(DMSO-d 6) 8: 0.83 (s, 3H), 1.15 (d,J 7.1 Hz, 3H), 1.27 (d,J 7.1 Hz, 3H), 1.35 (d,J 6.8 Hz, 4H), 1.48 (s, 3H), 1.55 1.70 (m, 3H), 1.77 1.88 (m, lH), 1.99 (d,J 13.5 Hz, lH), 2.12 (td,J 12.3, 6.6 Hz, Method r, lH), 2.28 2.40 (m, 3H), 2.48 (p,J 1.8 Hz, lH), 2.56 2.69 (m, lH), Table 7 3.59 (t,J 7.3 Hz, 2H), 4.06 4.28 (m, 3H), 4.33 (p,J 7.2 Hz, lH), 87 4.45 (dd,J 19.4, 6.4 Hz, lH), 4.85 (d,J 4.9 Hz, lH), 4.93 (t,J 7.2 Rt 0.72 min; Hz, lH), 5.03 (t,J 6.0 Hz, lH), 5 .28 (s, lH), 5.40 (dd,J 4.4, 1.9 Hz, m/z 911.64 lH), 6.02 (d,J 2.1 Hz, lH), 6.22 (dd,J 10.1, 1.9 Hz, lH), 6.43 (d,J [M+H+] 8.6 Hz, lH), 6.97 (s, 2H), 7.16 (d,J 7.8 Hz, lH), 7.20 7.33 (m, 4H), 7.44 7.49 (m, 2H), 7.90 (d,J 2.3 Hz, lH), 8.03 (d,J 7.3 Hz, lH), 8.17 (d,J 7.1 Hz, lH), 9.71 (s, lH) 1H NMR d 6) 8: 0.84 (s, 3H), 1.02 (ddd,J 21.3, 12.1, 4.2 Hz, 2H), 1.15 (d,J 7.1 Hz, 3H), 1.26 (d,J 7.1 Hz, 3H), 1.37 (s, 3H), 1.53 1.81 (m, 4H), 2.00 (dd,J 12.2, 5.5 Hz, lH), 2.04 2.15 (m, lH), 2.23 Method r, 2.33 (m, lH), 2.38 (dd,J 8.0, 6.6 Hz, 2H), 2.51 (d,J 18.2 Hz, lH), Table 7 3.59 (t,J 7.3 Hz, 2H), 3.87 (s, 2H), 4.16 (d,J 19.4 Hz, lH), 4.21 (p,J 88 7.1 Hz, lH), 4.27 (q,J 3.3 Hz, lH), 4.32 (p,J 7.1 Hz, lH), 4.48 (d, Rt 0.87 min; J 19.4 Hz, lH), 4.72 (s, 2H), 4.90 (d,J 5.4 Hz, lH), 5.37 (s, lH), 5.90 m/z 863.32 (t,J l.6 Hz, lH), 6.13 (dd,J 10.1, 1.8 Hz, lH), 6.88 (dt,J 7.9, 1.3 [M+H+] Hz, lH), 6.96 (s, 2H), 7.14 7.22 (m, 3H), 7.29 (d,J 10.1 Hz, lH), 7.33 7.38 (m, 2H), 7.43 (dd,J 7.8, 1.1 Hz, 2H), 8.03 (d,J 7.2 Hz, lH), 8.17 (d,J 7.0 Hz, lH), 9.71 (s, lH) 1H NMR (MeOH-d 4) 8: 1.01 (d,J 8.5 Hz, 3H), 1.10 1.30 (m, 2H), 1.34 (dd,J 10.5, 7.1 Hz, 3H), 1.45 (dd,J 7.2, 3.6 Hz, 3H), 1.52 (s, Methodm, 3H), 1.80 (t,J 13.0 Hz, lH), 1.90 (p,J 8.3, 7.3 Hz, lH), 2.04 (d,J Table 7 12.4 Hz, lH), 2.12 2.32 (m, 2H), 2.42 (d,J 11.2 Hz, lH), 2.46 2.58 (m, 2H), 2.60 2.78 (m, lH), 3.68 3.84 (m, 2H), 3.96 (d,J 6.0 Hz, Rt 1.99 min; 2H), 4.12 (d,J 19.3 Hz, lH), 4.16 4.27 (m, lH), 4.27 4.38 (m, lH), m/z 863 4.43 (d,J 6.2 Hz, 2H), 5.40 (d,J 6.3 Hz, lH), 6.05 (s, lH), 6.12 (d,J [M+H+] 4.4 Hz, lH), 6.28 (dd,J 9.9, 1.8 Hz, lH), 6.75 (d,J 3.3 Hz, 2H), 6.96 (d,J 7.7 Hz, lH), 7.15 7.30 (m, 5H), 7.43 (d,J 18.7 Hz, 3H), 7.56 (d,J 8.2 Hz, 3H) 1H NMR (DMSO-d 6) 8: 0.79 (d,J 6.8 Hz, 3H), 0.82 (d,J 6.8 Hz, 3H), 0.84 (s, 2H), 0.95 1.12 (m, 2H), 1.26 (d,J 7.1 Hz, 3H), 1.38 (d,J 4.7 Hz, 3H), 1.54 1.77 (m, 4H), 1.91 (h,J 6.8 Hz, lH), 1.96 2.05 Method r, (m, lH), 2.04 2.17 (m, lH), 2.23 2.34 (m, lH), 2.37 2.47 (m, 2H), Table 7 2.49 2.58 (m, lH), 3.51 3.67 (m, 2H), 3.87 (s, 2H), 4.11 (s, lH), 4.16 90 (d,J 19.4 Hz, lH), 4.27 (q,J 3.4 Hz, lH), 4.32 (p,J 7.1 Hz, lH), Rt 0.91 min; 4.48 (d,J 19.4 Hz, lH), 4.73 (s, lH), 4.90 (d,J 5.3 Hz, lH), 5.38 (s, m/z 891.36 lH), 5.90 (d,J 1.6 Hz, lH), 6.13 (dt,J 10.1, 1.8 Hz, lH), 6.88 (dt,J [M+H+] 7.7, 1.3 Hz, lH), 6.96 (s, 2H), 7.18 (dd,J 16.7, 8.1 Hz, 3H), 7.29 (d,J .1 Hz, lH), 7.32 7.40 (m, 3H), 7.43 (ddd,J 8.1, 2.2, 1.0 Hz, lH), 7.99 (d,J 8.4 Hz, lH), 8.10 (d,J 7.0 Hz, lH), 9.74 (s, lH) 1HNMR(DMSO-d 6) 8: 0.83 (s, 3H), 1.13 (d,J 7.1 Hz, 3H), 1.24 (d,J 7.1 Hz, 3H), 1.35 (qd, J 13.3, 12.8, 5.1 Hz, lH), 1.46 (s, 3H), 1.63 (q,J 1.88 (m, lH), 2.01 (dt, J 13.7, 3.5 Hz, lH), Method r, 9.7, 8.5 Hz, 3H), 1.73 2.14 (td,J 11.8, 7.2 Hz, lH), 2.26 2.40 (m, 3H), 2.48 2.69 (m, 2H), Table 7 3.57 (t,J 7.3 Hz, 2H), 3.85 (s, 2H), 4.17 (ddd,J 17.5, 11.7, 6.2 Hz, 99 3H), 4.30 (p,J 7.2 Hz, lH), 4.47 (d,J 19.4 Hz, lH), 4.83 4.95 (m, Rt 0.85 min; lH), 5.40 (s, 2H), 5.99 (d,J 1.6 Hz, lH), 6.20 (dd,J 10.1, 1.9 Hz, m/z 881.46 lH), 6.87 (d,J 7.5 Hz, lH), 6.95 (s, 2H), 7.16 (t,J 7.9 Hz, lH), 7.20 [M+H+] (d, J 8.1 Hz, 2H), 7.25 (d,J 10.1 Hz, lH), 7.31 (d,J 8.0 Hz, 2H), 7.38 (d,J 1.9 Hz, lH), 7.43 (dd,J 8.0, 2.0 Hz, lH), 8.01 (d,J 7.3 Hz, lH), 8.14 (d,J 7.1 Hz, lH), 9.70 (s, lH) 1H NMR (DMSO-d 6) 8: 0.93 (s, 3H), 1. 15 (d,J 7.2 Hz, 3H), 1.26 (d,J 7.1 Hz, 3H), 1.48 (s, 4H), 1.73 (dd,J 25.4, 11.3 Hz, 3H), 2.00 (d,J Method r, 14.1 Hz, lH), 2.12 2.26 (m, lH), 2.27 (s, lH), 2.37 (q,J 8.3, 7.8 Hz, Table 7 2H), 2.65 (d,J 33.3 Hz, lH), 3.59 (t,J 7.3 Hz, 2H), 3.88 (s, 2H), 4.14 4.27 (m, 2H), 4.32 (t,J 7.2 Hz, lH), 4.92 (d,J 3.5 Hz, lH), 5.50 (s, Rt 0.87 min; lH), 5.55 (s, lH), 5.54 5.72 (m, lH), 5.79 6.04 (m, 2H), 6.11 (s, lH), m/z 933.0 6.28 (dd,J 10.1, 1.9 Hz, lH), 6.90 (d,J 7.7 Hz, lH), 6.97 (s, 2H), [M+H+] 7.18 (t,J 7.8 Hz, lH), 7.24 (t,J 9.5 Hz, 3H), 7.34 (d,J 7.8 Hz, 2H), 7.40 (s, lH), 7.46 (d,J 8.2 Hz, lH), 8.03 (d,J 7.3 Hz, lH), 8.16 (d,J 7.1 Hz, lH), 9.72 (s, lH) DMSO-d 6) 8: 0.99 (s, 3H), 1.15 (d,J 7.1 Hz, 3H), 1.25 (d,J 7.1 Hz, 3H), 1.49 (s, 4H), 1.60 1.75 (m, 2H), 1.79 (d,J 14.0 Hz, lH), Method r, 1.94 (dt,J 14.4, 3.5 Hz, lH), 2.20 (q,J 10.4 Hz, lH), 2.24 2.33 (m, lH), 2.38 (dd,J 8.0, 6.5 Hz, 2H), 2.62 (<ltd, J 30.0, 12.0, 11.5, 4.1 Hz, Table 7 lH), 3.59 (t,J 7.3 Hz, 2H), 3.87 (s, 2H), 4.16 (d,J 9.1 Hz, lH), 4.21 101 (p, J 7.2 Hz, lH), 4.32 (p,J 7.2 Hz, lH), 5.00 (t,J 2.9 Hz, lH), 5.40 Rt 0.84 min; .47 (m, lH), 5.48 (s, lH), 5.54 5.72 (m, lH), 6.11 (s, lH), 6.27 (dd,J m/z 885.41 .2, 1.9 Hz, lH), 6.89 (d,J 7.6 Hz, lH), 6.96 (s, 2H), 7.17 (t,J 7.9 [M+H+] Hz, lH), 7.21 (d,J 8.0 Hz, 2H), 7.24 (dd,J 10.3, 1.5 Hz, lH), 7.32 (d, J 7.9 Hz, 2H), 7.39 (t,J 1.9 Hz, lH), 7.46 (dd,J 8.1, 2.1 Hz, lH), 8.04 (d,J 7.3 Hz, lH), 8.16 (d,J 7.1 Hz, lH), 9.73 (s, lH) 1HNMR(DMSO-d 6) 8: 0.85 (s, 3H), 1.16 (d,J 7.1 Hz, 3H), 1.28 (d,J 7.1 Hz, 3H), 1.48 (s, 3H), 1.56 (p,J 12.4, 12.0 Hz, lH), 1.62 1.76 (m, Method r, 3H), 1.98 2.10 (m, lH), 2.22 (td,J 12.3, 6.6 Hz, lH), 2.24 2.33 (m, Table 7 lH), 2.38 (td,J 7.0, 1.0 Hz, 2H), 2.54 2.72 (m, lH), 3.59 (t,J 7.3 Hz, 2H), 4.15 4.25 (m, 3H), 4.33 (p,J 7.1 Hz, lH), 4.50 (d,J 19.4 Rt 0.77 min; Hz, lH), 4.93 (d,J 5.0 Hz, lH), 5.36 (s, lH), 5.50 (s, lH), 5.55 5.73 m/z 917.22 (m, lH), 6.11 (q,J 1.5 Hz, lH), 6.27 (dd,J 10.2, 1.9 Hz, lH), 6.78 [M+H+] 6.83 (m, 2H), 6.84 (d,J 1.8 Hz, 2H), 6.97 (s, 2H), 7.01 (d,J 1.6 Hz, lH), 7.25 (dd,J 10.1, 1.5 Hz, lH), 7.45 7.61 (m, 2H), 8.04 (d,J 7.3 Hz, lH), 8.17 (d,J 7.0 Hz, lH), 9.73 (s, lH) 1HNMR(DMSO-d 6) 8: 0.84 (s, 3H), 1.15 (d,J 7.3 Hz, 3H), 1.25 (d,J 7.1 Hz, 3H), 1.48 (s, 3H), 1.51 (d,J 7.3 Hz, 4H), 1.67 (d,J 14.2 Hz, Method r, 2H), 2.02 (d,J 13.7 Hz, lH), 2.13 2.34 (m, 2H), 2.38 (t,J 7.3 Hz, Table 7 2H), 2.65 (s, lH), 3.59 (t,J 7.3 Hz, 2H), 4.08 (d,J 7.1 Hz, lH), 4.12 4.24 (m, 2H), 4.32 (t,J 7.2 Hz, lH), 4.48 (dd,J 19.6, 6.3 Hz, lH), Rt 0.84 min; 4.92 (d,J 4.8 Hz, lH), 5.07 (t,J 5.8 Hz, lH), 5.72-5.54 (m, lH), 5.41 m/z 935.4 (s, lH), 5.49 (s, lH), 6.11 (s, lH), 6.28 (d,J 9.9 Hz, lH), 6.88 6.95 [M+Na+] (m, lH), 6.97 (s, 2H), 7.17 (t,J 7.9 Hz, lH), 7.25 (d,J 8.1 Hz, 3H), 7.33 (d,J 7.9 Hz, 2H), 7.45 (d,J 12.0 Hz, 2H), 8.03 (d,J 7.2 Hz, lH), 8.16 (d,J 6.9 Hz, lH), 9.71 (s, lH) 1HNMR(DMSO-d 6) 8: 0.85 (s, 3H), 1.16 (d,J 7.1 Hz, 3H), 1.26 (d,J 7.1 Hz, 3H), 1.49 (s, 3H), 1.52 (d,J 7.3 Hz, 3H), 1.69 (t,J 12.7 Hz, 3H), 2.04 (d,J 13.9 Hz, lH), 2.18 2.33 (m, 2H), 2.33 2.42 (m, 2H), Method r, 2.56 2.74 (m, lH), 3.60 (t,J 7.3 Hz, 2H), 4.09 (q,J 7.2 Hz, lH), Table 7 4.20 (tq,J 13.0, 6.3, 5.6 Hz, 3H), 4.32 (p,J 7.1 Hz, lH), 4.49 (dd,J 104 19.5, 6.3 Hz, lH), 4.93 (d,J 5.1 Hz, lH), 5.08 (t,J 6.0 Hz, lH), 5.42 Rt 0.84 min; (s, lH), 5.51 (d,J 4.2 Hz, lH), 5.64 (dt,J 48.9, 8.9 Hz, lH), 6.12 (s, m/z 935.4 lH), 6.29 (dd,J 10.0, 1.9 Hz, lH), 6.93 (d,J 7.6 Hz, lH), 6.97 (s, [M+Na+] 2H), 7.18 (t,J 7.9 Hz, lH), 7.23 7.29 (m, 3H), 7.34 (d,J 8.2 Hz, 2H), 7.44 (d,J 2.1 Hz, lH), 7.44 7.49 (m, lH), 8.03 (d,J 7.3 Hz, lH), 8.17 (d,J 7.1 Hz, lH), 9.72 (s, lH) 1H NMR d 6) 8: 0.84 (s, 3H), 1.15 (d,J 7.2 Hz, 3H), 1.26 (d,J 7.1 Hz, 3H), 1.44 1.58 (m, 7H), 1.67 (d,J 13.8 Hz, 2H), 2.03 (d, J Method r, 14.0 Hz, lH), 2.18 2.34 (m, 2H), 2.38 (t,J 7.3 Hz, 2H), 2.50 2.72 Table 7 (m, lH), 3.58 (t,J 7.3 Hz, 2H), 4.07 (p,J 7.3 Hz, lH), 4.11 4.26 (m, 105 2H), 4.33 (d,J 7.2 Hz, lH), 4.48 (d,J 19.3 Hz, lH), 4.92 (d,J 4.8 Rt 0.80 min; Hz, lH), 5.41 (s, lH), 5.45 5.53 (m, lH), 5.62 (dd,J 48.5, 9.8 Hz, m/z 913.75 lH), 6.11 (s, lH), 6.28 (d,J 10.0 Hz, lH), 6.97 (s, 2H), 7.11 7.20 (m, [M+H+] 2H), 7.20 7.29 (m, 3H), 7.32 (d,J 8.0 Hz, 2H), 7.47 (d,J 8.3 Hz, 2H), 8.03 (d,J 7.2 Hz, lH), 8.16 (d,J 7.0 Hz, lH), 9.71 (s, lH) 1HNMR(DMSO-d 6) 8: 0.84 (s, 3H), 1.15 (d,J 7.1 Hz, 3H), 1.26 (d,J 7.1 Hz, 3H), 1.48 (s, 3H), 1.48 1.54 (m, 4H), 1.60 1. 75 (m, 2H), 2.03 (dt,J 13.8, 3.6 Hz, lH), 2.08 2.18 (m, lH), 2.18 2.25 (m, lH), 2.25 Method r, 2.32 (m, lH), 2.38 (dd,J 8.1, 6.4 Hz, 2H), 2.54 2.72 (m, lH), 3.59 Table 7 (t,J 7.3 Hz, 2H), 4.06 (dq, J 14.9, 7.0 Hz, lH), 4.11 4.28 (m, 3H), 106 4.33 (p,J 7.1 Hz, lH), 4.48 (d,J 19.6 Hz, lH), 4.92 (d,J 5.0 Hz, Rt 0.84 min; lH), 5.42 (s, lH), 5.45 5.54 (m, lH), 5.54 5.73 (m, lH), 6.12 (d,J m/z 913.46 2.2 Hz, lH), 6.26 6.33 (m, lH), 6.97 (d,J 1.3 Hz, 2H), 7.10 7.18 [M+H+] (m, 3H), 7.20 7.28 (m, 3H), 7.29 7.36 (m, 2H), 7.47 (dd,J 8.6, 4.4 Hz, 2H), 8.04 (d,J 7.3 Hz, lH), 8.17 (d,J 7.1 Hz, lH), 9.71 (d,J 2.9 Hz, lH) 1HNMR(DMSO-d 6) 8: 0.84 (s, 3H), 0.94 1.11 (m, 2H), 1.16 (d,J 7.1 Hz, 3H), 1.27 (d,J 7.1 Hz, 3H), 1.37 (s, 3H), 1.51 1.82 (m, 5H), 1.94 Method r, 2.03 (m, lH), 2.08 (d,J 19.3 Hz, lH), 2.30 (t,J 8.2 Hz, lH), 2.38 (t, Table 7 J 7.3 Hz, 2H), 2.48 2.59 (m, lH), 3.60 (t,J 7.3 Hz, 2H), 4.11 4.25 107 (m, 4H), 4.27 (d,J 3.6 Hz, lH), 4.32 (p,J 7.2 Hz, lH), 4.48 (d,J Rt 0.88 min; 19.5 Hz, lH), 4.90 (d, J 5.0 Hz, lH), 5.38 (s, lH), 5.91 (d,J 1.6 Hz, m/z 895.30 lH), 6.14 (dd,J 10.1, 1.8 Hz, lH), 6.97 (s, 2H), 6.94 7.01 (m, lH), [M+H+] 7.19 (t,J 8.0 Hz, lH), 7.25 7.37 (m, 2H), 7.37 (s, 4H), 7.66 (t,J 1.9 Hz, lH), 8.10 (d,J 7.2 Hz, lH), 8.20 (d,J 6.9 Hz, lH), 9.80 (s, lH) 1HNMR(DMSO-d 6) 8: 0.85 (s, 3H), 1.16 (d,J 7.1 Hz, 3H), 1.27 (d,J 7.1 Hz, 3H), 1.37 (qd, J 12.8, 5.2 Hz, lH), 1.48 (s, 3H), 1.57 1.71 (m, Method r, 3H), 1.83 (dt,J 11.7, 5.4 Hz, lH), 2.03 (dt,J 13.8, 3.6 Hz, lH), 2.14 Table 7 (td,J 12.1, 6.8 Hz, lH), 2.30 2.36 (m, lH), 2.38 (t,J 7.3 Hz, 2H), 108 2.42 2.57 (m, lH), 2.63 (td,J 13.3, 5.8 Hz, lH), 3.59 (t,J 7 .3 Hz, Rt 0.83 min; 2H), 4.13 4.26 (m, 5H), 4.32 (p,J 7.2 Hz, lH), 4.49 (d,J 19.4 Hz, m/z 913.0 lH), 4.91 (d,J 4.7 Hz, lH), 5.42 (s, 2H), 6.02 (s, lH), 6.21 (dd,J [M+H+] 10.1, 1.9 Hz, lH), 6.98 (dd, J 7.9, 1.8 Hz, lH), 7.19 (t,J 8.0 Hz, lH), 7.27 (d,J 10.2 Hz, lH), 7.29 7.44 (m, 5H), 7.61 7.70 (m, lH), 8.07 (d, J 7.2 Hz, lH), 8.18 (d,J 7.0 Hz, lH), 9.79 (s, lH) 1HNMR(DMSO-d 6) 8: 0.85(s,3H),1.19 (d,J 7.1 Hz,3H), 1.28(d,J 7.1 Hz, 3H), 1.49(s,4H),1.61 1.78(m,3H), 2.04(d,J 13.7 , 2.22(dd,J 12.3,6.3 Hz,lH), 2.26 2.32(m,lH),2.37(td,J 7.0, 2.0 Method r, Hz, 2H),2.55 2.73(m,lH),3.58(t,J 7.3 Hz, 2H), 3.78 (s, 2H),4.18 Table 7 (d,J 19.4 Hz, lH), 4.29(p,J 7.1 Hz,lH), 4.39 (p,J 7.1 Hz,lH), 109 4.50(d,J 19.4 Hz,lH), 4.93(d,J 5.2 Hz,lH), 5.43(s,lH),5.51(dd, Rt 0.80 min; J 4.5, 1.7 Hz, lH),5.54 , lH),6.12(s,lH),6.29(dd,J 10.1, m/z 915.54 1.9 Hz, lH),6.72 6.78(m,2H), 6.98(s,2H), 7.19 (d,J 7.9 Hz,2H), [M+H+] 7.25(dd,J 10.1, 1.5 Hz,lH), 7.29 7.37(m,2H), 7.76 (d,J 1.8 Hz, lH),8.18(d,J 7.5 Hz, lH),8.30(d,J 7.1 Hz, lH), 8.92 (s, lH),9.72 (s,lH) 1H NMR (DMSO-d 6) 8: 0.83(s,3H),1.13(d,J 7.2 Hz,3H), 1.25(d,J 7.2 Hz, 3H), 1.36 (qd,J 12.7, 5.4 Hz,lH), 1.46 (s,3H),1.55 1.72(m, Method r, 3H),1.75 1.87(m,lH), 1.93 2.05(m,lH), 2.05 2.18(m,lH),2.25 Table 7 2.40(m,3H),2.47(p,J 1.9 Hz,lH),2.53 2.69(m,lH),3.56(t,J 110 7.3 Hz, 2H), 4.09 4.23(m, 3H),4.29(p,J 7.1 Hz, lH),4.49 (d,J Rt 0.84 min; 19.5 Hz, lH), 4.87 ,lH),5.40(s,lH),5.43(s,lH),5.99(d,J m/z 899.0 1.7 Hz, lH),6.19 (dd,J 10.1,1.9 Hz,lH),6.95(s,2H),7.00(dt,J [M+H+] 7.8,1.3 Hz, lH), 7.21 7.33(m,4H),7.33 7.41(m,2H),7.56(ddd,J 8.2,2.1,1.0 Hz, lH), 7.67(t,J 2.0 Hz,lH), 8.05 (d,J 7.1 Hz,lH), 8.14(d,J 7.0 Hz,lH),9.87(s,lH) 1H NMR (DMSO-d 6) 8: ,3H), 1.04(ddd,J 14.7,11.7,4.1 Hz, 14(d,J 7.1Hz,3H),l.26(d,J 7.1 Hz,3H),l.37(s,3H),1.54 Method r, 1.85(m,4H),1.93 2.03(m, lH), 2.09 (d,J 11.6 Hz,lH),2.23 Table 7 2.33(m,lH), 2.36(q,J 6.4,5.5 Hz, 2H),2.50(d,J 9.7 Hz,lH),3.57 111 (t,J 7.3 Hz, 2H), 4.12 4.24(m,2H),4.24 4.34(m,2H),4.48(d,J Rt 0.86 min; 19.5 Hz, lH), 4.76(s,lH), 4.90(d,J 5.1 Hz,lH),5.39(s,lH),5.89(s, m/z 881.38 lH),6.14 (dd,J 10.1,1.9 , 6.97 (s,2H), 7.03(d,J 7.7 Hz, [M+H+] lH), 7.22 7.35(m,4H),7.42(d,J 8.2 Hz,2H),7.57(dd,J 8.2, 2.0 Hz, lH), 7.71 (d,J 2.0 Hz,lH),8.09(d,J 7.1 Hz,lH), 8.17 (d,J 6.9 Hz, lH),9.89(s,lH) 1H NMR (DMSO-d 6) 8: 0.84(s,3H), 1.02(ddd,J 21.5,11.7,4.1 Hz, 2H),1.18(d,J 7.1 Hz, 3H),1.27(d,J 7.1 Hz, 3H),1.37(s, 3H), 1.54 1.81(m,5H), 1.95 2.03(m, lH), 2.09 (dd,J 11.1,4.0 Hz,lH), 2.29 Method r, (d,J 12.8 Hz, lH), 2.36(td,J 7.1, 2.0 Hz, 2H), 2.49 ,lH), Table 7 3.57(d,J 7.3 Hz,2H), 3.77(s,2H),4.15(d,J 19.4 Hz,lH), 4.23 112 4.34(m,2H), 4.38 (p,J 7.1 Hz,lH),4.47(d,J 19.5 Hz,lH),4.74(s, Rt 0.85 min; lH),4.89(d,J 5.3 Hz, lH),5.37(s,lH),5.90(t,J 1.6 Hz,lH),6.13 m/z 879.30 (dd,J 10.1, 1.9 Hz, lH),6.73(d,J 1.8 Hz,2H), 6.97(s,2H), 7.17 (d, [M+H+] 2H),7.28 (d,J 10.1 Hz,lH),7.31 ,2H), 7.76 (d,J 1.5 Hz, 17 (d,J 7.6 Hz, lH), ,J 7.1 Hz, lH), 8.90 (s, lH),9.72 (s,lH) 1H NMR (DMSO-d 6) 8: 0.84(s,3H), 1.04(ddd,J 1.8,4.1 Hz, 2H),1.14 (d,J 7.1 Hz, 3H), 1.25(d,J 7.1 Hz, 3H), 1.38 (s, 3H), 1.54 1.81(m,4H), 1.95 2.04(m, lH), 2.10(tt,J 10.9,5.9 Hz, lH), 2.25 Method r, 2.33(m,lH),2.37(dd,J 8.0,6.5 Hz, 49 2.59(m,lH),3.58(t, Table 7 J 7.3 Hz, 2H), 3.78 (s, 2H), 4.15(d,J 19.4 Hz,lH),4.21(p,J 7.1 113 Hz, lH), 4.25 4.36 (m,2H),4.45(d,J 19.4 Hz, lH), 4.76 (s, lH),4.87 Rt 0.80 min; (d,J 5.3 Hz, 27(s,lH),5.92(t,J 1.6 Hz, lH), 6.14 (dd,J m/z 880.26 .1,1.9 Hz,lH), 6.77(dd,J 7.7,1.6 Hz, 84 6.91(m,2H), [M+H+] 6.97(s,2H),6.99(d,J 7.7 ,7.11 7.17(m,lH),7.30(d,J .1 Hz, lH), 7.39 7.45(m,2H),8.03(d,J 7.3 Hz, lH),8.16(d,J 7.2 Hz, lH),9.55(s,lH),9.72(s,lH) Example 36: Synthesis of minophenyl)((6aR 6bS, 7S, 8aS, 8bS, l0R llaR, 12aS, 12bS) hydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyloxo-2, 4, 6a, 6b, 7, 8, 8a, 8b, l la, 12, 12a, 12bdodecahydro-1H-naphtho [2', l':4, 5]indeno[l, 2-d][l, 3]dioxolyl)benzamide Step 1: Synthesis of 4-Formylbenzoyl chloride Oxalyl chloride (17.51 mL, 200 mmol) was added drop-wise to a 0 °C solution of 4- formylbenzoic acid (15.01 g, 100 mmol) in THF (100 mL), followed by N,N-dimethylformamide (0.387 mL, 5.00 mmol) in drop-wise addition. The mixture was allowed to warm to room temperature and then stirred for an additional 2 h. The mixture was concentrated in vacuo to give ylbenzoyl chloride (16.86 g, 100 mmol, 100 % yield), which was used without further purification.
Step 2: Synthesis of tert-Butyl (3-(4-formylbenzamido )phenyl)carbamate BocHN CHO 'r("y N v NH2 H Cl BocHN N � YY PCHO 0 v a 9] Triethylamine (63.4 mL, 455 mmol) was added drop-wise to a 0 °C solution of 4- formylbenzoyl de (16.86 g, 100 mmol) in THF (100 mL), followed by addition of tert-butyl (3- aminophenyl)carbamate (18.93 g, 91 mmol). After stirring at room temperature for 2 h, the mixture was diluted with EtOAc (200 mL), washed with water (2 x 100 mL) and brine (100 mL), dried over Na2SO4, and concentrated in vacuo. The residue was triturated in 20 mL of EtOAc/PE (1:1), and the resulting precipitate was collected to give tert-butyl (3-(4-formylbenzamido)phenyl)carbamate (27.8 g, 82 mmol, 90% yield) as a yellow solid. LCMS (Method e Table 7) R1 2.00 min; MS m/z 285 [M-t-Bu].
Step 3: Synthesis of N-(3-Aminophenyl)((6aR 6bS, 7S, 8aS, 8bS, l0R, l laR, 12aS, 12bS)hydroxy-8b-(2-hydroxyacetyl)-6a, ethyloxo-2, 4, 6a, 6b, 7, 8, 8a, 8b, l la, 12, 12a, 12bdodecahydro-1H-naphtho [2', l':4, 5]indeno[l, 2-d][l, 3]dioxolyl)benzamide To a d solution of (8S, 9S, l0R, llS, 13S, 14S, 16R l, 16, 17-trihydroxy (2- hydroxyacetyl)-10, 13-dimethyl-6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, l7-dodecahydro-3H­ cyclopenta[a]phenanthrenone (9.42 g, 25.02 mmol), tert-butyl (3-(4-formylbenzamido)phenyl) carbamate , made in a manner similar to Example 2, Step 5, (8.515 g, 25.02 mmol and MgSO4 (12.04 g, 100 mmol) in MeCN(250 mL) was added drop-wise trifluoromethanesulfonic acid (11.11 ml, 125 mmol) at 0 °C. The mixture was stirred at 0 °C for 2 hours and then warmed to room ature and stirred for additional 16 h. The mixture was ed and washed with THF, and the filtrate was concentrated in vacuo. The residue was dissolved in THF (100 mL) and then neutralized with 1 M NaOH aqueous solution to pH 8. The e was extracted with EtOAc (200 mL), washed with water (2 x 100 mL) and brine (100 mL), dried over Na2SO4, and trated in vacuo. Purification by chromatography (silica) eluting with 5% MeOH/DCM gave crude product, which was further purified by reverse phase HPLC on a Sunfire Cl8 10 micron (250 x 19 mm column). A gradient of MeCN (A) and 0.05% TFA in water (B) was used, at a flow rate of 30 mL/min (0-10.0 min linear gradient22-32%, hold 5 min). Combined fractions were frozen and lyophilized to give N-(3-aminophenyl)((6aR 6bS, 7S, 8aS, 8bS, l0R, l laR 12aS, 12bS)hydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyloxo-2, 4, 6a, 6b, 7, 8, 8a, 8b, Ila, 12, 12a, 12b-dodecahydro-1H-naphtho[2', l':4, 5]indeno[l, , 3]dioxolyl)benzamide (1.972 g, 3.29 mmol, 13% yield) as a white solid. LCMS (Method f, Table 7) R1 1.37 min; MS m/z +]. 1H 599 [M+H NMR (400 MHz, Methanol-d4) 8 8.01 - 7.92 (m, 3H), 7.64 (d, J 8.0 Hz, 2H), 7.55 -7.40 (m, 3H), 7.05 (d, J= 7.8 Hz, lH), 6.27 (dd, J= 10.2, 1.8 Hz, lH), 6.03 (s, lH), 5.60 (s, lH), 5.13 (d, J= 4.1 Hz, lH), 4.68 (d, J 19.4 Hz, lH), 4.45 (d, J 3.3 Hz, lH), 4.37 (d, J 19.4 Hz, lH), 2.68 (dt, J 14.5, 7.0 Hz, lH), 2.41 (dd, J= 13.7, 10.2 Hz, lH), 2.29 (d, J= 10.5 Hz, lH), 2.18 (d, J= 12.8 Hz, lH), 1.99 (dd, J= 13.8, 3.5 Hz, lH), 1.94 -1.80 (m, 2H), 1.82 -1.69 (m, 2H), 1.52 (s, 3H), 1.14 (m, J 16.8, 8.0 Hz, 2H), 1.02 (s, 3H).
Minor acetal isomer: N-(3-Aminophenyl)((6aR,6bS,7S,8aS,8bS,10S, llaR12aS,12bS) hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b, 7 ,8,8a,8b, l la, 12, 12a, 12b-dodecahydro- 1H-naphtho[2',l ':4,5]indeno[ l ,2-d][ oxol- l enzamide (112 mg, 0.176 mmol, 0.9% yield) as a yellow solid. LCMS (Method e, Table 7) R1 1.53 min; MS m/z +]. 1H NMR (400 MHz, 599 [M+H MeOH-d4) 8 7.90 (d, J 7.9 Hz, 2H), 7.48 (dd, J 8.1, 3.5 Hz, 3H), 7.15 (d, J 2.2 Hz, lH), 7.09 (t, J 8.0 Hz, lH), 6.96 (d, J 8.1 Hz, lH), 6.59 - 6.51 (m, lH), 6.28 (dd, J 10.1, 1.9 Hz, lH), 6.25 (s, lH), 6.05 (s, lH), 5.51 -5.37 (m, lH), 4.45 (s, lH), 4.30 (d, J 19.2 Hz, lH), 4.14 (d, J 19.2 Hz, lH), 2.70 (t, J 13.6 Hz, lH), 2.43 (d, J 13.3 Hz, lH), 2.22 (dd, J 23.3, 12.5 Hz, 2H), 2.07 (d, J 13.5 Hz, lH), 1.93 (q, J 5.1, 3.5 Hz, 2H), 1.80 (d, J 14.0 Hz, 2H), 1.53 (d, J 1.7 Hz, 3H), 1.21 (dd, J 41.7, 12.1 Hz, 2H), 1.03 (s, 3H).
Example 37: Synthesis ofN-(3-Aminophenyl)((6aS,6bR7S,8aS,8bS, l0R,l laR,12aS,12bS)-6b-fluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro- 1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol-l 0-yl)benzamide Synthesized using the same procedure as Example 36 above. Major acetal isomer: N-(3- Aminophenyl)((6aS,6bR,7S,8aS,8bS,1 OR l aS,12bS)-6b-fluoro- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl oxo-2,4,6a,6b,7,8,8a,8b,l la,12, 12a, 12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)benzamide. LCMS (Method f, Table 7) Rt l.35 min; MS m/z +]. 1H NMR (400 MHz, DMSO-d 617 [M+H 6) 8 10.29 (d, J 7.4 Hz, lH), 8.00 - 7.93 (m, 2H), 7.73 (s, lH), 7.59 (d, J 8.1 Hz, 2H), 7.43 (s, OH), 7.38 (s, lH), 7.29 (dd, J 10.0, 5.3 Hz, 2H), 6.81 (s, lH), 6.24 (dd, J 10.1, 1.9 Hz, lH), 6.05 (d, J 1.6 Hz, lH), 5.62 (s, lH), 5.49 (s, lH), 5.03 -4.96 (m, lH), 4.58 (d, J 19.5 Hz, lH), 4.23 (d, J 19.6 Hz, lH), 2.73 -2.52 (m, lH), 2.40 -2.32 (m, lH), 2.25 - 2.12 (m, lH), 2.11 -2.02 (m, lH), 1.92 - 1.84 (m, lH), 1.76 -1.67 (m, 3H), 1.51 (s, 3H), 1.40 (tt, J 14.3, 7.1 Hz, lH), 0.90 (s, 3H).
Minor acetal isomer: N-(3-Aminophenyl)((6aS,6bR7S,8aS,8bS,1 OS, llaR12aS,12bS)-6bfluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a, 6b, 7,8,8a,8b, 11a,12,12a, 12bdodecahydro-1H-naphtho [2',l ':4,5]indeno[ l ,2-d][ l,3]dioxol-l0-yl)benzamide. LCMS (Method B, Table 7) R1 1.45 min; MS m/z +]. 1H NMR (400 MHz, DMSO-d 617 [M+H 6) 8 ppm: 8.06 (s, lH), 7.95 (d, J 8.4 Hz, 2H), 7.54-7.41 (m, 5H), 7.09 (d, J 6.8 Hz, lH), 6.34 (d, J 10 Hz, lH), 6.28 (s, lH), 6.13 (s, lH), 5.49 (d, J 6.4 Hz, lH), 4.34-4.13 (m, 3H), 2.79-2.24 (m, 5H), 1.74-1.63 (m, 2H), 1.60 (s, 3H), 1.04 (s, 3H).
Example 38: Synthesis ofN-(3-Aminophenyl)((2S,6aS,6bR7S,8aS,8bS,lOR,l laR,12aS,12bS)-2,6b­ difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la,12, 12a, 12bdodecahydro-1H-naphtho [2',l':4,5]indeno[l,2-d] [l,3]dioxol-l0-yl)benzamide E E O O a�SS ' -P r!t!P o, .. r!t!P HO•• j S : 0 2; � o OH O OH HO''' OH n_f _ _ , � (',,··· � + , N I : � T H � O H H N � 0 MgS04 vI Y""" v OH I OH OH ; 0 0 Synthesized using the same procedure as Example 36 above. Major acetal isomer: N-(3- Aminophenyl)((2S,6aS,6bR,7S,8aS,8bS, 1 OR llaR,12aS, 12bS)-2,6b-difluoro- 7-hydroxy-8b-(2- yacetyl)-6a,8a-dimethyl -2,4,6a,6b,7,8,8a,8b, l la,12, 12a, 12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)benzamide. LCMS (Method f, Table 7) R1 1.376 min, MS m/z +]. 1H NMR (400 MHz, MeOH-d 635 [M+H 4) 8 7.96 (d, J 8.0 Hz, 2H), 7.88 (d, J 2.1 Hz, lH), 7.64 (dd, J 8.3, 1.5 Hz, 2H), 7.48 -7.42 (m, lH), 7.39 (t, J 7.9 Hz, lH), 7.34 (d, J 10.0 Hz, lH), 6.98 (dt, J 7.7, 1.6 Hz, lH), 6.41 - 6.26 (m, 2H), 5.71 -5.45 (m, 2H), 5.14 (d, J 4.1 Hz, lH), 4.69 (d, J 19.4 Hz, lH), 4.44 -4.28 (m, 2H), 2.73 (dt, J 25.9, 12.1 Hz, lH), 2.41 (td, J 11.7, 6.9 Hz, 2H), 2.29 (dt, J 14.0, 3.6 Hz, lH), 1.91 - 1.67 (m, 4H), 1.60 (s, 4H), 1.02 (s, 3H).
Minor acetal : N-(3-Aminophenyl)((2S,6aS,6bR,7S,8aS,8bS,1OS,llaR,12aS,12bS)- 2,6b-difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dim 4-oxo-2,4,6a,6b,7,8,8a,8b,lla, 12,12a, 12b- hydro-1H-naphtho[2',l':4,5]indeno[l,2-d][l,3]dioxolyl)benzamide. LCMS (Method e, Table 7) Rt 1.506 min, MS m/z 635 [M+ H +]. 1HNMR (400 MHz, MeOH-d 4) 8 7.90 (d, J= 8.0 Hz, 2H), 7.49 (d, J 7.9 Hz, 2H), 7.35 (d, J 10.0 Hz, lH), 7.16 (d, J 2.2 Hz, lH), 7.09 (t, J 8.0 Hz, lH), 6.96 (d, J 8.2 Hz, lH), 6.65 6.48 (m, lH), 6.41 6.27 (m, 3H), 5.76 5.39 (m, 2H), 4.42 4.22 (m, 2H), 4.15 (d, J 19.4 Hz, lH), 2.66 (dd, J 27.8, 13.4 Hz, lH), 2.47 2.24 (m, 3H), 2.08 1.85 (m, 2H), 1.75 (t, J 14.9 Hz, 2H), 1.61 (s, 3H), 1.03 (s, 3H).
Example 39: Synthesis of3-Aminophenyl 4-((2S,6aS,6bR,7S,8aS,8bS,1OR,l laR,12aS,12bS)-2,6b­ difluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, 12a, ecahydro-1H-naphtho [2', l':4,5]indeno[l,2-d][l,3]dioxol-l 0-yl)benzoate Step 1: Synthesis of tert-Butyl (3-hydroxyphenyl)carbamate (BochO I THF H2N v OH BocHN OH 85 oc, 16 h 'r(Y 'r(Y v To a solution of 3-aminophenol (10 g, 92 mmol) in THF( 450 mL) was added Boe anhydride (23.40 mL, 101 mmol). The mixture was heated at 85 °C for 16 h, monitored by LCMS. After that, the mixture was concentrated to obtain a residue, which was dissolved in EtOAc (150 mL) and washed with water (100 mL), saturatedNaHCO 3 (100 mL) and brine (100 mL), dried overNa2SO4, and concentrated in vacuo. The crude al was washed with PE (50 mL x 2) to give the title compound (16.5 g, 76 mmol, 82% yield) as a white solid. LCMS (Method g, Table 7) Rt 1.66 min, MS m/z 232.1 [M+Na+].
Step 2: Synthesis of 3-((tert-Butoxycarbonyl)amino)phenyl 4-formylbenzoate CHO BocHN',("y v o H CHO (Y N HO� BocHN o� DCC,DMAP yy DCM, rt, 16 h v o To a solution of utyl (3-hydroxyphenyl)carbamate (5 g, 23.90 mmol) in DCM (60 mL) at 0 °C was added 4-formylbenzoic acid (3.59 g, 23.90 mmol), N, N'-dicyclohexylcarbodiimide (7.40 g, .8 mmol) and thylaminopyridine (0.584 g, 4.78 mmol). The resulting mixture was d at that temperature for 10 min under an atmosphere of argon. Then the mixture was warmed to room temperature and stirring was continued for 16 h. The mixture was cooled in an ice bath. The side product N, N'-dicyclohexylurea was filtered off as a precipitate and the clear filtrate was concentrated under vacuum. The crude material was purified by silica gel chromatography eluting with DCM/ EtOAc (100%-30:1) to give the title compound (7.0 g, 18.54 mmol, 78% yield) as a white solid. LCMS (Method d, Table 7) Rt 2.l 7 min, MS m/z 364.0 [M+Na +].
Step 3: Synthesis of 3-Aminophenyl ,6aS,6bR7S,8aS,8bS,1OR l laR 12aS,12bS)-2,6bdifluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, 12a, 12bdodecahydro-1H-naphtho [2', l':4,5]indeno[l,2-d][l,3]dioxol- l 0- yl)benzoate To a d solution of (6S, 8S, 9R l0S, llS, 13S, 14S, 16R 17S)-6 , 9-difluoro- 11, 16, 17- trihydroxy(2-hydroxyacetyl)-10, 13-dimethyl-6 , 7, 8, 9, 10, 11, 12, 13, 14 , 15, 16, l7-dodecahydro- 3H-cyclopenta[a]phenanthrenone (Step 5, Example 2) (7.248 g, 17.58 mmol) and 3-((tertbutoxycarbonyl )amino)phenyl yl benzoate (6 g, 17.58 mmol) in anhydrous MeCN (40 mL) and THF (40 mL) at 0 °C under nitrogen was added ise triflu oromethanesulfonic acid (7.8 mL, 87.9 mmol). The mixture was stirred at 0 °C for 1 h, then poured onto ice water (30 mL) and extracted with EtOAc (2 x 45 mL). The combined organic layers were washed with cooled water (2 x 30 mL), brine (30 mL), ted NaHCO 3 (30 mL) and additional water (30 mL), concentrated in vacuo affording a yellow solid. The crude material was purified by silica gel column chromatography (200-300 mesh), eluting with DCM/ MmeOH (100%-40:1) and then further purified by prep HPLC to give the title compound (major acetal isomer) (2.166 g, 3.32 mmol, 19% yield) as a white solid. LCMS (Method d, Table 7) R1 1.54 min ; MS m/z +]. 1H NMR (400 MHz, DMSO-d 636.3 [M+H 6) 8 8.13 (d, J= 8.2 Hz, 2H), 7.66 (d, J= 8.2 Hz, 2H), 7.27 (dd, J 10.1 , 1.4 Hz, lH), 7.06 (t, J 8.0 Hz, lH), 6.48 (dd, J 8.3 , 2.1 Hz, lH), 6.40 (t, J 2.2 Hz, lH), 6.34 (dd, J 7.8, 2.2 Hz, lH), 6.30 (dd, J 10.1 , 1.9 Hz, lH), 6.12 (s, lH), 5.82- .47 (m, 3H), 5.31 (s, 2H), 5.15 (t, J= 5.9 Hz, lH), 5.03 (d, J= 5.1 Hz, lH), 4.60 (dd, J= 19.5, 6.4 Hz, lH), 4.33-4.12 (m, 2H), 2.66 (ddd, J 26.2 , 13.7 , 9.3 Hz, lH), 2.31 (d, J 10.9 Hz, lH), 2.21 (td, J 12.4, 6.3 Hz, lH), 2.12 - 1.98 (m, lH), 1.84-1.64 (m, 3H), 1.50 (s, 4H), 0.89 (s,3H). 3-Aminophenyl 4-((2S, 6aS, 6bR, 7S, 8aS, 8bS, l0S, llaR 12aS, 12bS)-2, luoro-7 - hydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyloxo-2, 4, 6a, 6b, 7, 8, 8a, 8b, l la, 12, 12a, 12bdodecahydro-1H-naphtho [2', l':4 , 5] indeno [l,2-d][l,3]dioxolyl)benzoate, the minor acetal isomer was also isolated (1.073 g, 1.676 mmol, 10% yield) as a white solid. LCMS (Method d, Table 7) R1 1.58min; MSm/z +]. 1HNMR (400 MHz, DMSO-d 636.3 [M+H 6) 8 8.13- 8.06 (m, 2H), 7.51 (d,J 8.2 Hz,2H), 7.28 (dd, J 10.1 , 1.4 Hz, lH), 7.06 (t, J 8.0 Hz, lH), 6.52- 6.44 (m, lH), 6.43- 6.28 (m, 4H), 6.14 (s, lH), 5.80 - 5.48 (m, 2H), 5.39 (d, J 6.8 Hz, lH), 5.31 (s, 2H), 5.04 (t, J 6.1 Hz, lH), 4.26 - 4.15 (m, 2H), 4.05 (dd,J 19.2 , 5.9 Hz, lH), 2.67 - 2.51 (m, lH), 2.29 (d, J 6.9 Hz, lH), 2.27- 2.14 (m, lH), 2.11 (d,J= 13.5 Hz, lH), 1.96 - 1.59 (m, 4H), 1.51 (s, 3H), 0.90 (s,3H). e 40: Synthesis of3-Aminophenyl 4-((6aS,6bR,7S,8aS,8bS, I 0R,l laR,12aS, 12bS)-6b-fluoro- 7- hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,Ila,12,12a,12b-dodecahydro- htho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)benzoate aminophenyl 4-((6aS,6bR,7S,8aS,8bS,I OR, llaR,12aS,12bS)-6b-fluoro- 7-hydroxy-8b-(2-hydroxyacetyl)- 6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,I la,12, 12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d][l,3]dioxol-l0-yl)benzoate. LCMS (Method d, Table 7) R1 1.54 min; MS m/z +]. 1H 618.3 [M+H NMR (400 MHz, MeOH-d4) 8 8.19 (dd, J 7.8, 1.9 Hz, 2H), 7.67 (d, J 8.0 Hz, 2H), 7.41 (d, J IO.I Hz, IH), 7.35 (t, J 8.0 Hz, IH), 6.97 -6.84 (m, 3H), 6.32 (dd, J IO. I, 1.9 Hz, IH), 6.12 (s, IH), 5.63 (s, IH), 5.13 (d, J 5.0 Hz, IH), 4.69 (d, J 19.4 Hz, IH), 4.43- 4.30 (m, 2H), 2.76 (td, J 13.6, 5.8 Hz, IH), 2.70-2.54 (m, IH), 2.43 (d, J= 13.6 Hz, IH), 2.31 (ddd, J= 14.9, 11.6, 4.5 Hz, 2H), 2.01- 1.92 (m, IH), 1.89 - 1.69 (m, 3H), 1.62 (s, 4H), 1.03 (s, 3H).
Minor acetal isomer: 3-aminophenyl 4-((6aS,6bR,7S,8aS,8bS,l0S,llaR,12aS,12bS)-6bfluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a, 6b, 7,8,8a,8b,11a,12,12a, ecahydro-1H-naphtho [2',l':4,5]indeno[ l ,2-d][ l ,3]dioxol-l0-yl)benzoate. LCMS (Method d, Table 7) R1 1.58 min; MS m/z +]. 1H NMR (400 MHz, DMSO-d 618.2 [M+H 6) 8 8.10 (d, J 8.1 Hz, 2H), 7.52 (d, J 8.1 Hz, 2H), 7.30 (d, J IO. I Hz, IH), 7.16 (dd, J 9.8, 6.0 Hz, IH), 6.64 (d, J 8.2 Hz, IH), 6.61 -6.51 (m, 2H), 6.31-6.19 (m, 2H), 6.05 (s, IH), 5.47 (s, lH), 5.38 (d, J= 6.7 Hz, lH), 4.19 (d, J= 18.7 Hz, 2H), 4.04 (d, J 19.2 Hz, lH), 2.66 (td, J 13.9, 6.3 Hz, lH), 2.48 - 2.33 (m, lH), 2.17-2.05 (m, 2H), 1.87 (dt, J 13.8, 7.0 Hz, 2H), 1.84 - 1.69 (m, 2H), 1.51 (s, 4H), 0.90 (s, 3H).
Example 41: Synthesis of 3-Aminophenyl 4-((6aR,6bS,7S,8aS,8bS,l0R,llaR,12aS,12bS)hydroxy-8b­ (2-hydroxyacetyl)-6a,8a- dimethyl oxo-2,4,6a,6b,7,8,8a,8b, lla,12, b-dodecahydro-1H­ naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)benzoate 0 0 ,,. + H,N O H,N O OH yv��OH I ... I 0 ,,,, 0 V V Synthesized using the same ure as Example 39 above. Major acetal isomer: 3- aminophenyl 4-((6aR,6bS,7S,8aS,8bS,1OR, llaR,12aS,12bS)hydroxy-8b-(2-hydroxyacetyl)-6a,8adimethyl oxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d][l,3]dioxol-l0-yl)benzoate. LCMS (Method d, Table 7) Rt l.86 min; MS m/z +]. 1H 599.8 [M+H NMR (400 MHz, MeOH-d4) 8 8.14 (d, J= 8.2 Hz, 2H), 7.64 (d, J= 8.1 Hz, 2H), 7.44 (d, J= 10.0 Hz, lH), 7.14 (t, J 8.0 Hz, lH), 6.63 (dd, J 8.1, 2.2 Hz, lH), 6.54 (q, J 2.6 Hz, lH), 6.48 (dd, J 8.1, 2.2 Hz, lH), 6.24 (dd, J 10.0, 2.0 Hz, lH), 6.00 (s, lH), 5.59 (s, lH), 5.13 (d, J 4.4 Hz, lH), 4.69 (d, J 19.4 Hz, lH), 4.43 (q, J 3.3 Hz, lH), 4.37 (d, J 19.4 Hz, lH), 2.66 (td, J 13.4, 5.3 Hz, lH), 2.38 (dd, J 13.7, 4.1 Hz, lH), 2.32 - 2.19 (m, lH), 2.14 (d, J 12.7 Hz, lH), 2.06 - 1.93 (m, lH), 1.94 - 1.85 (m, lH), 1.89 - 1.68 (m, 3H), 1.50 (s, 3H), 1.22 - 1.01 (m, 2H), 1.02 (s, 3H).
Minor acetal isomer: 3-aminophenyl 4-((6aR,6bS,7S,8aS,8bS,l0S, l laR,12aS,12bS) hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7 ,8,8a,8b, lla,12,12a,12b-dodecahydro- 1H-naphtho[ 2',l':4,5]indeno[l,2-d][l,3]dioxol-l0-yl)benzoate. LCMS d d, Table 7) Rt l.89 min; MS m/z +]. 1HNMR (400 MHz, MeOH-d 599.8 [M+H 4) 8 8.22 - 8.11 (m, 2H), 7.53 (d, J= 8.2 Hz, 2H), 7.49 (d, J 10.0 Hz, lH), 7.14 (t, J 8.0 Hz, lH), 6.63 (dd, J 8.1, 2.1 Hz, lH), 6.55 (t, J 2.2 Hz, lH), 6.53 - 6.45 (m, lH), 6.33 -6.24 (m, 2H), 6.05 (t, J 1.6 Hz, lH), 5.46 (t, J 3.8 Hz, lH), 4.46 (q, J 3.3 Hz, lH), 4.30 (d, J= 19.2 Hz, lH), 4.15 (d, J= 19.2 Hz, lH), 2.70 (td, J= 13.5, 5.4 Hz, lH), 2.48 - 2.38 (m, lH), 2.23 (ddd, J 24.3, 12.6, 6.6 Hz, 2H), 2.12 -2.03 (m, lH), 2.02 - 1.89 (m, 2H), 1.89 - 1.77 (m, 2H), 1.53 (s, 3H), 1.26 (tt, J 12.4, 6.3 Hz, lH), 1.17 ( dd, J 11.1, 3.6 Hz, lH), 1.03 (s, 3H).
Example 42: Synthesis of(6aR,6bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((3- Aminophenoxy)methyl)pheny1)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphth o[2', l ':4,5]indeno[l,2-d] ioxolone 7] Step 1: Synthesis of tert-Butyl (3-( myl benzyl)oxy)phenyl)carbamate r(YC HO BocHN OH CHO U r-7T{ BrvV BocHN O� K2C03, DMF vI K2CO3 (47.8 g, 346 mmol) and tert-butyl (3-hydroxyphenyl)carbamate (36.2 g, 173 mmol) were added sequentially to a solution of 4-(bromomethyl)ben zaldehyde (34.4 g, 173 mmol) in dimethyl ide (200 mL). The yellow suspension was then heated at 80 °C in an oil bath for 2 h. The reaction was quenched with water (200 mL) and extracted with EtOAc (2 x 200 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, and were concentrated under reduced pressure. The residue obtained was purified by chromatography (silica gel; petroleum ether to 80:20 PE/EtOAc; gradient elution) to provide tert-butyl (3-((4-formyl benzyl)oxy)phenyl)carbamate (47.27 g, 144 mmol, 83% yield) as a white solid. LCMS (Method h Table 7) R1 1.92 min; MS m/z +]. 272 u+H 9] Step 2: Synthesis of (6aR,6bS,7S,8aS,8bS, l0R, l laR,12aS,12bS)(4-((3- Aminophenoxy)methyl)pheny1)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b, 7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d] [l,3]dioxolone Trifluoromethanesulfonic acid (17. 76 mL, 200 mmol) was added drop-wise to a stirred 0 °C sion of (8S,9S,10R,l 1S,13S,14S,16R, l7S)- l l,16, l7-trihydroxy-l 7- (2-hydroxyacetyl)-10,13- dimethyl-6,7,8,9,10,11,12,13,14,15,16,l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (15.06 g, 40 mmol) and tert-butyl -formylbenzyl)oxy)phenyl) carbamate (13.75 g, 42.0 mmol), and MgSO4 (19.26 g, 160 mmol) in MeCN (400 mL). The reaction mixture was then warmed to 20 °C and stirred for an additional 2 h. The mixture was filtered and washed with THF, and the filtrate was concentrated in vacuo. The residue was dissolved in THF (100 mL), neutralized with 1 M aqueous NaOH to pH 8, d with EtOAc (200 mL), washed with water (2 x 200 mL) and brine (200 mL), dried (Na 2SO4), and concentrated in vacuo. The residue was purified by flash column (MeOH:DCM l:20). The resulting material was purified further by prep HPLC on a Sunfire Cl8 10 micron (250 x 19 mm column). A gradient of MeCN (A) and 0.05% TFA in water (B) was used, at a flow rate of 30 mL/min (0-10.0 min linear gradient 22-32% A, hold 5 min) to give the title compound (7.338 g, 12.15 mmol, 30% yield) as a yellow solid. LCMS (Method i, Table 7) R1 1.47 min; MS m/z 1H NMR (400 MHz, 586 [M+H +].
MeOD-d4) 8 7.502-7.446 (m, 5H), 7.389-7.349 (m, lH), 7.009, 6.988 (dd, JI= 2 Hz, J2 8.4 Hz, lH), 6.890-6.859 (m, 2H), 6.275, 6.250 (dd, JI 1.2 Hz, J2 8.8 Hz, lH), 6.027 (s, lH), 5.501 (s, lH), 5.147 (s, 2H), 5.107, 5.078 (dd, JI 6.8 Hz, J2 11.6 Hz, lH), 4.672 (d, J 19.6 Hz, lH), 4.436 (s, lH), 4.370 (d, J 19.2 Hz, lH), 2.706-2.671 (m, lH), 2.265 (m, 3H), 1.700 (m, 5H), 1.512 (s, 3H), 1.112 (m, lH), 1.054-1.009 (m, 4H).
Minor acetal isomer: (6aR,6bS,7S,8aS,8bS,1OS,llaR,12aS,12bS)(4-((3- aminophenoxy)methyl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl - l ,2,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H-naphtho[2',l ':4,5]indeno[l,2-d][l,3]dioxolone, the minor acetal isomer was also isolated (354 mg, 0.604 mmol, 2% yield) as a yellow solid. LCMS (Method i, Table 7) R1 1.51 min; MS m/z +]. 1H NMR (400 MHz, DMSO-d 586 [M+H 6) 8 7.416 (d, J 8 Hz, 2H), 7.332-7.276 (m, 3H), 6.879 (t, J 8 Hz, lH), 6.185-6.115 (m, 5H), 5.948 (s, lH), 5.319 (d, J 6.8 Hz, lH), 5.041-5.014 (m, 3H), 4.980 (s, 2H), 4.791 (d, J 3.2 Hz, lH), 4.302-4.239 (m, 2H), 4.056, 4.008 (dd, JI 6 Hz, J2 19.6 Hz, lH), 2.552-2.540 (m, lH), 2.337-2.304 (m, lH), 2.075-2.005 (m, 2H), 1.884-1.736 (m, 5H), 1.395 (s, 3H), 1.219-1.045 (m, 2H), 0.892 (s, 3H).
Example 43: sis of(6aS,6bR,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((3- aminophenoxy)methyl)phenyl)-6b-fluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b, 7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxolone , ,�,,v,__,,er '" ' ' r+f+P'' H 8 2 �� OH OH � + � � I TfOH H, N 0»--·· H, N O ff MgSO4 OH v OH OH vI I sized using the same procedure as Example 42 above. Major acetal isomer: (6aS,6bR,7S,8aS,8bS,1 OR, l laR, 12aS,12bS)- l 0-(4-((3-aminophenoxy)methyl)phenyl)-6b-fluoro- 7- hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H­ naphtho[2',l':4,5]indeno[l,2-d][l,3]dioxolone. LCMS (Method i, Table 7) Rt=l .74 min; MS m/z = 604 [M+H +]. 1H NMR (400 MHz, CD 3OD) 8 7.48 - 7.49 (m, 4H), 7.44-7.33 (m, 2H), 7.02 (dd, J= 8.3, 1.9 Hz, lH), 6.96- 6.84 (m, 2H), 6.32 (dd,J= 10.1, 1.8 Hz, lH), 6.13 (s, lH), 5.52 (s, lH), 5.16 (s, 2H), 5.08 (d, J= 4.9 Hz, lH), 4.65 (d, J= 19.4 Hz, lH), 4.46 - 4.27 (m, 2H), 2.84-2.50 (m, 2H), 2.45- 2.27 (m, 3H), 2.01 -1.90 (m, lH), 1.80- 1.70 (m, 3H), 1.62 (s, 3H), 1.55 (dd, J=12.8, 4.8 Hz, lH), 1.02 (s, 3H).
Minor acetal isomer: (6aS,6bR,7S,8aS,8bS,1 OS, l laR,12aS,12bS)(4-((3- aminophenoxy)methyl)phenyl)-6b-fluorohydroxy -8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la, 12, 12a, 12b-dodecahydro-4H-naphth o[2', l ':4,5]indeno[l,2-d][l,3]dioxolone.
LCMS (Method i, Table 7) Rt=l.77 min; MS m/z = 604 [M+H +].1H NMR (400 MHz, DMSO) 8 7.42 (d, J=8.1 Hz, 2H), 7.36 -7.25 (m, 3H), 7.01 (t, J= 8.1 Hz, lH), 6.43 - 6.30 (m, 3H), 6.24 (dd, J= 10.1, 1.5 Hz, lH), 6.12 (s, lH), 6.04 (s, lH), 5.47 (s, lH), 5.35 (d, J 7.1 Hz, lH), 5.02 (s, 2H), 4.31- 4.14 (m, 2H), 4.04 (d, J = 19.2 Hz, lH), 2.72 -2.58 (m, lH), 2.18-1.98 (m, 2H), 1.85 (d, J= 6.9 Hz, 2H), 1.77 - 1.63 (m, 2H), 1.58 - 1.40 (m, 4H), 0.90 (s, 3H).
Example 44: Synthesis of(2S,6aS,6bR 7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l 0-( 4-( (3- Aminophenoxy )methyl)phenyl )-2, 6b-difluo ydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyll ,2, 6a, ,8a,8b, l la,12,12a, 12b-dodecahydro-4H-naphth o[2', l indeno[l,2-d][l,3]dioxolone CHO 0 � O BocHN O Ho,.. rrW f!.��· H� OH "' l _ »,,• T OH H,N yy O MgS04 OH �yy��V OH OH V Synthesized usmg the same procedure as Example 42 above. Major acetal isomer: (2S,6aS,6bR,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((3-aminophenoxy)methyl)phenyl)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H­ naphtho[2',l':4,5]indeno[l,2-d][l,3]dioxolone. LCMS (Method f, Table 7) Rt=l.45 min; MS m/z = 622 [M+H +].1H NMR (400 MHz, DMSO) 8 7.44 (s, 4H), 7.27 (d, J= 10.1 Hz, lH), 6.87 (t, J= 8.0 Hz, lH), 6.30 (dd, J I 0.1, 1.5 Hz, lH), 6.24 - 6.04 (m, 4H), 5.81 -5.39 (m, 3H), 5.13 (t, J 5.9 Hz, lH), 5.09 - 4.91 (m, 5H), 4.55 (dd, J 19.5, 6.4 Hz, lH), 4.32 - 4.09 (m, 2H), 3.60 (t, J 6.3 Hz, 2H), 2.81 - 2.55 (m, lH), 2.40 - 2.14 (m, 2H), 2.06 (d, J 13.6 Hz, lH), 1.85 -1.63 (m, 6H), 1.58 - 1.43 (m, 4H), 0.88 (s, Minor acetal isomer: (2S,6aS,6bR,7S,8aS,8bS,1OS, l laR 12aS,12bS)-l 0-( 4-( (3- aminophenoxy)methyl)phenyl)-2, 6b-difluo rohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2, 6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H-naphth o[2', l ':4,5]indeno[l,2-d][l,3]dioxolone.
LCMS (Method f, Table 7) R1 1.49 min; MS m/z +]. 1HNMR (400 MHz, DMSO) 8 7.41 (d, 622 [M+H J 8.1 Hz, 3H), 7.35 - 7.20 (m, 3H), 6.88 (t, J 8.0 Hz, lH), 6.31 (dd, J 10.1, 1.6 Hz, lH), 6.16 (dd, J 13.9, 5.0 Hz, 6H), 5.77 - 5.45 (m, 2H), 5.36 (d, J 7.1 Hz, lH), 4.35 -4.13 (m, 2H), 4.05 (dd, J 18.9, 4.9 Hz, lH), 2.70 - 2.53 (m, lH), 2.29 (s, lH), 2.24 -2.13 (m, lH), 2.12 - 2.04 (m, lH), 1.96 - 1.81 (m, lH), 1.81 - 1.63 (m, 3H), 1.50 (s, 4H), 0.89 (s, 3H).
Example 45: Synthesis of bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((3- Aminobenzyl)oxy)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyll ,2, 6a, 6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxolone Step 1: Synthesis of tert-Butyl (3-(hydroxymethyl)phenyl)carbamate H2N OH __(B_oc_h_o_ BocHN OH lf lf To a solution of (3-aminophenyl)methanol (88.5 g, 719 mmol) in THF (80 mL) was added di­ tert-butyl dicarbonate (184 mL, 790 mmol). The mixture was stirred at 25 °C overnight. The mixture was then concentrated to s and the residue was purified by silica gel column chromatography (eluted with EtOAc/hexanes l:9, v/v) to afford the title compound (161.1 g, 722 mmol, 100% yield), as a white solid. LCMS (Methodj, Table 7) R1 1.77 min; MS m/z p 2: Synthesis of tert-Butyl (3- 246 [M+Na methyl)phenyl)carbamate H2N�v NBS, PPh3 BocHN� OH v Br To a on of tert-butyl (3-(hydroxymethyl)phenyl)carbamate (120 g, 484 mmol) in THF (50 mL) at -20 °C was added triphenylphosphine (254 g, 967 mmol) followed by N-bromosuccinimide (103 g, 580 mmol). After stirring for 3 h, the solvent was removed in vacuo, and the residue was purified by silica gel column chromatography (eluted with hexane:EtOAc 100:1) to provide the title compound (125 g, 437 mmol, 90% yield) as a white solid. LCMS dj, Table 7) R1 2.10 min; MS m/z 230, 232 [M-t-Bu+H+].
Step 3: Synthesis of tert-Butyl (3-( (4-formylphenoxy)methyl)phenyl)carbamate i""'Y �CHO BocHN HO� BocHNVA)J VI o :::,,,. I A e of 4-hydroxybenzaldehyde (25.6 g, 210 mmol) and potassium carbonate (29.0 g, 210 mmol) in dimethyl formamide (300 mL) was stirred for 15 min. Then tert-butyl (3- (bromomethyl)phenyl)carbamate (60 g, 210 mmol) was added. The mixture was heated to 60 °C and stirred for 2 hours at this temperature. The mixture was poured into 50 mL ofwater, extracted with EtOAc (3 x 50 mL). The combined organic layers was washed with water (lxl00 mL) and brine (lxl00 mL), concentrated in vacuum. The crude material was purified by silica gel column tography ( eluted with dichloromethane/methanol 500: 1) to afford the title compound (72 g, 209 mmol, 100% yield) as a white solid. LCMS dj, Table 7) R1 2.08 min; MS m/z +]. 272 [M-t-Bu+H Step 4: Synthesis of (6aR,6bS, 7S,8aS,8bS, l0R l laR12aS,12bS)(4-((3- aminobenzyl)oxy)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2, 6a, 6b,7,8,8a,8b, l la, 12, 12a, 12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone o 0 f"""Y BocHNVN O o,..
✓,; Lo,,.• TfOH H2N ,0•••· MgSO4 O I OH 2] Trifluoromethanesulfonic acid (12.38 mL, 139 mmol) was added drop-wise to a mixture of magnesium sulfate (13.43 g, 112 mmol), utyl (3-((4-formyl phenoxy)methyl)phenyl)carbamate (10.96 g, 33.5 mmol) and (8S,9S,10R,11S,13S,14S,16Rl7S)- l l, 16, l7-trihydroxy(2-hydroxyacetyl)- , 13-dimethyl-6,7,8,9, 10,11,12,13,14,15,16,l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (10.5 g, 27.9 mmol) in MeCN (150 mL) at 00C. The mixture was warmed to room temperature and stirred for 2 hours at this temperature. The mixture was filtered and the filtrate was poured into 500 mL of saturated sodium bicarbonate solution, extracted with EtOAc (250 mL). The organic layer was washed with brine (200 mL) and water (200 mL), concentrated in vacuo. The crude material was ed by silica gel column chromatography (eluted with DCM-MeOH 50: 1, v/v), and the ing product was purified further by prep-HPLC to afford the title compound (6.04 g, 10.31 mmol, 37% yield) as a white solid.
LCMS (Method k, Table 7) Rt l.91 min; MS m/z +]. 1HNMR (400 MHz, DMSO-d 586 [M+H 6) 8 7.35 (dd, J 19.6, 9.4 Hz, 3H), 7.09 6.89 (m, 3H), 6.61 6.33 (m, 3H), 6.18 (dd, J 10.1, 1.7 Hz, lH), 5.95 (s, lH), 5.38 (s, lH), 5.16 5.01 (m, 3H), 5.02 4.85 (m, 3H), 4.80 (d, J 3.0 Hz, lH), 4.50 (dd, J 19.5, 6.3 Hz, lH), 4.31 (s, lH), 4.18 (dd, J 19.4, 5.5 Hz, lH), 2.33 (d, J 10.5 Hz, lH), 2.17 1.98 (m, 2H), 1.90 1.53 (m, 5H), 1.40 (s, 3H), 1.13 0.96 (m, 2H), 0.87 (s, 3H).
Example 46: Synthesis of (6aS,6bR,7S,8aS,8bS, l0R,llaR,12aS,12bS)(4-((3- aminobenzyl)oxy)phenyl)-6b-fluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b, 7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone o CHO r-7l( BocHN }0 O 0'" HO"' VI h ,0,,,·L0, ... TfOH H2N MgS04 O OH I Synthesized using the same procedure as Example 45 above. LCMS (Method k, Table 7) R1 1.89 min; MS m/z +]. 1H NMR (400 MHz, DMSO-d 604 [M+H 6) 8 7.32 (dd, J= 17.3, 9.4 Hz, 3H), 7.10 - 6.94 (m, 3H), 6.65 - 6.35 (m, 3H), 6.25 (dd, J= 10.1, 1.7 Hz, lH), 6.05 (s, lH), 5.58 -5.32 (m, 2H), 5.22 - 5.03 (m, 3H), 5.01 - 4.86 (m, 3H), 4.52 (dd, J 19.5, 6.4 Hz, lH), 4.20 (dd, J 19.4, 5.5 Hz, 2H), 2.78 - 2.56 (m, lH), 2.44 - 2.31 (m, lH), 2.19 (td, J 12.0, 6.8 Hz, lH), 2.06 (d, J 13.7 Hz, lH), 1.95 - 1.81 (m, lH), 1.68 (dd, J= 15.4, 9.7 Hz, 3H), 1.57 - 1.30 (m, 4H), 0.88 (s, 3H).
Example 47: Synthesis of(2S,6aS,6bR,7S,8aS,8bS, l0R,l laR,12aS,12bS)(4-((3- enzyl)oxy)phenyl)-2,6b-difluo rohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la, 12, 12a,12b-dodecahydro-4H-naphtho[ 2', l':4,5]indeno[l,2-d][l,3]dioxolone 4] Synthesized using the same procedure as Example 45 above. LCMS (Method C, Table 7) R1 1.45 min; MS m/z +]. 1H NMR (400 MHz, CD 622 [M+H 3OD) 8 7.57 - 7.29 (m, 6H), 7.24 (d, J 7.7 Hz, lH), 7.02 (d, J 8.6 Hz, 2H), 6.45 -6.23 (m, 2H), 5.69 - 5.49 (m, lH), 5.46 (s, lH), 5.16 (s, 2H), .06 (d, J 3.7 Hz, lH), 4.64 (d, J 19.5 Hz, lH), 4.43 - 4.15 (m, 2H), 2.89 - 2.56 (m, lH), 2.52 - 2.32 (m, 2H), 2.28 (d,J 13.8 Hz, lH), 1.87 - 1.62 (m, 4H), 1.60 (s, 3H), 1.00 (s, 3H).
Minor acetal isomer: (2S,6aS,6bR,7S,8aS,8bS,1 0S,1laR,12aS,12bS)- l 0-( 4-( (3- aminobenzyl)oxy)phenyl)-2,6b-difluo rohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, ,8a,8b, l la, 12, 12a,12b-dodecahydro-4H-naphtho[ 2', l':4,5]indeno[l,2-d][l,3]dioxolone.
LCMS (Method C, Table 7) Rt l.48 min; MS m/z +]. 1H NMR (400 MHz, DMSO) 8 7.41 - 622 [M+H 7.13 (m, 3H), 7.08 -6.90 (m, 3H), 6.61 (s, lH), 6.52 (dd, J 17.3, 7.5 Hz, 2H), 6.31 (d, J 10.2 Hz, lH), 6.11 (d, J 18.4 Hz, 2H), 5.79 - 5.56 (m, lH), 5.53 (d, J 3.3 Hz, lH), 5.34 (d, J 7.2 Hz, lH), 5.18 - .00 (m, 3H), 4.93 (s, 2H), 4.28 (dd, J 19.1, 6.2 Hz, lH), 4.19 (d, J 5.9 Hz, lH), 4.05 (dd, J 19.1, 5.9 Hz, lH), 3 .60 (t, J 6.2 Hz, 3H), 2. 72 - 2.51 (m, lH), 2.29 (s, lH), 2.22 - 2.11 (m, lH), 2.06 (d, J 13 .4 Hz, lH), 1.93 - 1.80 (m, lH), 1.80 -1.60 (m, 6H), 1.50 (s, 3H), 1.36 (s, lH), 0.89 (s, 3H).
Example 48: Synthesis of(6aR,6bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((3- Aminophenyl)ethynyl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b, 7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxolone Step 1: Synthesis oftert-Butyl (3-ethyn ylphenyl)carbamate Boc2O H2N� THF BocHN�v Di-tert-butyl dicarbonate (123 ml, 531 mmol) was added to a stirred solution of 3- ethynylaniline (56.6 g, 483 mmol) in THF (300 mL). The mixture was heated to reflux for overnight. The mixture was then cooled to ambient temperature and taken up in ethyl acetate (500 mL) and washed sequentially with IN aqueous HCl (200 mL), saturated aqueous Na2CO3 (200 mL) and brine (200 mL).
The organic layer was dried over Na2SO4, concentrated in vacuo, and purified by silica gel column chromatography (eluted with 15% EtOAc/PE) to give tert-butyl (3-ethynylphenyl)carbamate (94 g, 435 mmol, 90% yield). LCMS (Method f, Table 7) Rt l.80 min; MS m/z +]. 162 [M-t-Bu+H Step 2: Synthesis -Butyl (3-( (4-formylphenyl)ethynyl)phenyl)carbamate BocHN�v IO Pd(PPh3}2Cl2 Cul, TEA, THF In a 500 mL of round bottom flask 4-iodobenzaldehyde (30.2 g, 130 mmol), bis(triphenylphosphine)palladium(II) chloride (4.56 g, 6.50 mmol), copper(!) iodide (2.476 g, 13.00 mmol) and triphenylphosphine (3.41 g, 13.00 mmol) were dissolved in THF (200 mL) and triethylamine (181 mL, 1300 mmol) followed by addition oftert-butyl (3-ethynylphenyl)carbamate (28.2 g, 130 mmol).
The mixture was stirred at 75 °C under nitrogen atmosphere for 16 h. After completion ofthe reaction, the volatile solvents were completely removed. The crude material was purified by silica gel column chromatography (eluted with PE/CH2Ch l:3) to obtain utyl (3-( ( 4- phenyl)ethynyl)phenyl)carbamate (35.5 g, 111 mmol, 85% yield) as an ite solid. LCMS (Method f, Table 7) R1 2.08 min; MS m/z +]. 322 [M+H 0] Step 3: Synthesis of (6aR,6bS,7S,8aS,8bS, l0R,l laR,12aS,12bS)(4-((3- Aminophenyl)ethynyl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b, ,8b, l la, 12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxoloneceo #f! o "�'"✓ MgS04 1] Trifluorometha onic acid (4.44 ml, 50.0 mmol) was added drop-wise to a 0 °C suspension of (8S,9S,10R,l1S,13S,14S,16R,17S)-ll, 16, l 7-trihydroxy (2-hydroxyacetyl)-10, 13- dimethyl-6, 7,8,9,10,11,12, 15,16, l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (3.76 g, .00 mmol), tert-butyl (3-((4-formylphenyl)ethynyl)phenyl) carbamate (3.21 g, 10 mmol) and MgSO4 (4.81 g, 40.0 mmol) in MeCN (100 ml). The mixture was stirred for additional 2 h. The e was filtered and washed with THF. The te was concentrated in vacuo. The residue was dissolved in THF (50 mL), neutralized with 1 M aqueous NaOH aqueous solution to pH 8, extracted with EtOAc (200 mL), washed with water (2 x 100 mL) and brine (100 mL), dried over (Na 2SO4, and trated in vacuo. The residue was purified by column chromatography (silica gel, eluted with MeOH : DCM 1 :40) to give 2.5 g of the crude product which was further purified by prep-HPLC to afford the title compound (1.449 g, 2.500 mmol, 25% yield) as a yellow solid. LCMS (Method 1, Table 7) R1 1.86 min; MS m/z 580 [M+H +]. 1H NMR (400 MHz, DMSO-d 6) 8 7.65 - 7.41 (m, 4H), 7.32 (d, J 9.9 Hz, 2H), 7.05 (dd, J 9.8, 5.7 Hz, lH), 6.85 - 6.49 (m, 4H), 6.29 -6.05 (m, lH), 6.01 -5.83 (m, lH), 5.63 - 5.40 (m, lH), 5.26 (s, 2H), .12 (t, J 5.8 Hz, lH), 4.96 (d, J 4.3 Hz, lH), 4.82 (d, J 3.0 Hz, lH), 4.63 - 4.41 (m, lH), 4.37 - 4.08 (m, 2H), 2.40 -1.91 (m, 5H), 1.87 - 1.52 (m, 6H), 1.40 (s, 4H), 1.14 -0.95 (m, 2H), 0.88 (s, 3H).
The minor acetal , (6aR, 6bS, 7S, 8aS, 8bS, l0S, llaR, 12aS, 12bS)(4-((3- aminophenyl)ethynyl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyl-6a, 6b, 7, 8, 8a, 8b, Ila, 12, 12a, 12b-decahydro-1H-naphtho[2', l':4, 5]indeno[ l, 2-d][ l, 3]dioxol-4(2H)-one, also was isolated (85 mg, 0.147 mmol, 1.5% yield) as a yellow solid. LCMS (Method i, Table 7) R1 1.93 min; MS m/z 580 [M+H +]. 1H NMR (400 MHz, DMSO-d 6) 8 7.51 (d, J 7.6 Hz, 2H), 7.42 - 7.21 (m, 3H), 7.05 (t, J 7.7 Hz, lH), 6.83 -6.45 (m, 3H), 6.29 -6.07 (m, 2H), 5.95 (s, lH), 5.47 -5.14 (m, 3H), 4.82 (s, lH), 4.38 - 4.14 (m, 2H), 4.03 (d, J= 19.3 Hz, lH), 2.33 (d, J= 10.3 Hz, 2H), 2.15 - 1.96 (m, lH), 1.93 - 1.68 (m, 5H), 1.40 (s, 3H), 1.33 -0.97 (m, 3H), 0.89 (s, 3H).
Example 49: Synthesis of (6aS,6bR,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((3- Aminophenyl)ethynyl)phenyl )-6b-fluorohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2, 6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[ l,2-d][l,3]dioxolone 0 a�,"✓ '' -�#f! , /o�#! MgSO4 � 00 � 00 A A 3] Synthesized using the same procedure as Example 48 above. Major acetal isomer: (6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l0-(4-((3-aminophenyl)ethynyl)phenyl)-6b-fluoro- 7- hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H­ naphtho[2',l':4,5]indeno[ l ,2-d][ l,3]dioxolone. LCMS (Method f, Table 7) R1 1.57 min; MS m/z 598 [M+H+]. 1H NMR (400 MHz, DMSO-d 6) 8 7.55 (d, J 8.1 Hz, 2H), 7.46 (d, J 8.1 Hz, 2H), 7.30 (d, J 10.1 Hz, lH), 7.05 (t, J 7.8 Hz, lH), 6.71 (s, lH), 6.66 (d, J 7.5 Hz, lH) , 6.60 (d, J 7.8 Hz, lH), 6.24 (d, J 8.9 Hz, lH), 6.04 (s, lH), 5.51 (d, J 15.2 Hz, 2H), 5.26 (s, 2H), 4.97 (d, J 4.4 Hz, lH), 4.55 (d, J 19.5 Hz, lH), 4.22 (d, J 19.5 Hz, 2H), 2.74 2.56 (m, lH), 2.36 (d, J 9.7 Hz, lH), 2.24 2.10 (m, lH), 2.06 (d, J 14.5 Hz, lH), 1.92 1.78 (m, lH), 1.78 1.58 (m, 3H), 1.50 (s, 3H), 1.45 1.31 (m, lH), 0.88 (s, 3H).
Minor acetal isomer: (6aS,6bR,7S,8aS,8bS,1OS, l laR,12aS,12bS)( 4-((3- aminophenyl)ethynyl)phenyl)-6b-fluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-4H-naphth o[2', l':4,5]indeno[l,2-d][l,3]dioxolone.
LCMS (Method f, Table 7) R1 1.61 min; MS m/z +]. 1HNMR (400 MHz, DMSO-d 598 [M+H 6) 8 7.51 (d, J 8.1 Hz, 2H), 7.39 7.22 (m, 3H), 7.05 (t, J 7.8 Hz, lH), 6.72 (s, lH), 6.67 (d, J 7.6 Hz, lH), 6.60 (d, J 8.1 Hz, lH), 6.24 (d, J 10.1 Hz, lH), 6.18 (s, lH), 6.05 (s, lH), 5.49 (s, lH), 5.35 (d, J 6.9 Hz, lH), 5.26 (s, 2H), 4.19 (d, J 18.9 Hz, 2H), 4.04 (d, J 19.1 Hz, lH), 2.75 2.55 (m, lH), 2.37 (d, J .2 Hz, lH), 2.09 (d, J 7.1 Hz, 2H), 1.84 (d, J 6.5 Hz, 2H), 1.78 1.62 (m, 2H), 1.50 (s, 4H), 0.89 (s, 3H).
Example 50: Synthesis of(2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l 0-(4-((3- Aminophenyl)ethynyl)phenyl)-2, 6b-difluorohydroxy- hydroxyacetyl)-6a,8a-dimethyl - l ,2,6a, 6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-4H-naphth o[2', l':4,5]indeno[l,2-d][l,3]dioxolone F � O BocHNif ,4#0, /0�# HQI• -------- MgS04 � 00 � 00 h h Synthesized usmg the same ure as Example 48 above. Major acetal isomer: S,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l 0-(4-((3-aminophenyl)ethynyl)phenyl)-2,6b-difluoro- 7- hydroxy-8b-(2-hydroxyacetyl) -6a,8a-dimethyl-l ,2,6a,6b,7,8,8a,8b, l 12a,12b-dodecahydro-4H­ naphtho[2',l ':4,5]indeno[ l,2-d][l,3]dioxolone. LCMS (Method f, Table 7) R1 1.57 min; MS m/z 616 [M+H+].1HNMR (400 MHz, DMSO-d 6) 8 7.55 (d, J 8.1 Hz, 2H), 7.47 (d, J 8.2 Hz, 2H), 7.27 (d, J 10.1 Hz, lH), 7.05 (t, J 7.8 Hz, lH), 6.71 (s, lH), 6.66 (d, J 7.6 Hz, lH), 6.60 (d, J 8.1 Hz, lH), 6.30 (dd, J= 10.1, 1.4 Hz, lH), 6.13 (s, lH), 5.80 5.58 (m, lH), 5.55 (d, J= 7.1 Hz, 2H), 5.26 (s, 2H), .14 (t, J 5.9 Hz, lH), 4.99 (d, J 5.1 Hz, lH), 4.56 (dd, J 19.5, 6.4 Hz, lH), 4.23 (dd, J 19.5, 5.4 Hz, 2H), 2.79 2.56 (m, lH), 2.31 (s, lH), 2.26 2.14 (m, lH), 2.12 1.99 (m, lH), 1.83 1.62 (m, 3H), 1.61 1.40 (m, 4H), 0.88 (s, 3H).
Minor acetal isomer: (2S,6aS,6bR,7S,8aS,8bS,1OS, l aS,12bS)- l 0-( 4-( (3- aminophenyl)ethynyl)pheny1)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-4H-naphth o[2', l':4,5]indeno[l,2-d][l,3]dioxolone.
LCMS (Method f, Table 7) R1 1.61 min; MS m/z +]. 1HNMR (400 MHz, DMSO-d 616 [M+H 6) 8 7.52 (d, J 8.3 Hz, 2H), 7.33 (d, J 8.2 Hz, 2H), 7.28 (d, J 10.0 Hz, IH), 7.05 (t, J 7.8 Hz, IH), 6.73 (s, IH), 6.68 (d, J 7.5 Hz, IH), 6.61 (d, J 8.1 Hz, IH), 6.32 (dd, J IO.I, 1.7 Hz, IH), 6.21 (s, IH), 6.15 (s, IH), 5.78 5.58 (m, IH), 5.55 (d, J 2.7 Hz, IH), 5.36 (t, J 7.9 Hz, IH), 5.27 (s, 2H), 5.08 (t, J .8 Hz, IH), 4.33 4.12 (m, 2H), 4.06 (dd, J 19.1, 5.0 Hz, IH), 2.72 2.53 (m, IH), 2.29 (s, IH), 2.23 2.02 (m, 2H), 1.92 1.82 (m, IH), 1.82 1.61 (m, 3H), 1.51 (s, 4H), 0.90 (s, 3H).
Example 51: Synthesis of(2S,6aS,6bR,7S,8aS,8bS,l0R, l laR,12aS,12bS)(4-((E) Aminosty ry1 )pheny 1 )-2, 6b-difluo rohydroxy-8b-(2-hydroxyacety1)-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b,Ila,12,12a,12b-dodecahydro-4H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxolone Step 1: sis of tert-Butyl (E)-(3-(2-(4,4,5,5-tetrameth yl-l,3,2-dioxaborolan yl)vinyl)phenyl)carbamate BocHN� j< BocHN � v RuH(PPh3l3(CO)CI v PhMe, 50 °C 4,4,5,5-Tetramethyl-l, 3, 2-dioxaborolane (16.70 mL, 115 mmol) and carbonylchlorohydridotris(triphenylphosphine)ruthenium(II) (2.63 g, 2.76 mmol) were added to a nitrogen-purged solution -butyl (3-ethynylphenyl)carbamate (10 g, 46.0 mmol) in toluene (150 mL).
The mixture was heated at 50 °C for 16 h, whereupon it was concentrated under reduced pressure.
Purification by chromatography (silica) eluting with PE/ EtOAc (100%-10:1) gave the title compound (13.25 g, 36.8 mmol, 80% yield) as a white solid. LCMS (Method d Table 7) R1 2.19 min; MS m/z 290.1 [M-tBut. 1H NMR (400 MHz, CDC1 3) 8 1.33 (s, 12H), 1.54 (s, 9H), 6.17 (d, J 18.4 Hz, lH), 6.49 (bs, lH), 7.18 (d, J 7.6 Hz, lH), 7.26-7.29 (m, lH), .40 (m, 2H), 7.47 (s, lH).
Step 2: Synthesis oftert-Butyl (E)-(3-(4-formylstyryl)phenyl)carbamate p BrPhCHO K2C03, Pd(PPh3)4 CHO j< THF, 80 °C - ,,,.__ � n BocHN � BocHNu "s/ "s/ To a on of tert-Butyl (E)-(3-(2-(4,4,5,5-tetrameth yl- l ioxaborolan yl)vinyl)phenyl)carbamate (6 g, 17.38 mmol) and 4-bromobenzaldehyde (3.38 g, 18.25 mmol) in THF (85 mL) at 20 °C under N2 were added K2CO3 (4.80 g, 34.8 mmol) and Pd(Ph3P)4 (1.607 g, 1.390 mmol). The e was stirred at 80 °C for 32 h. The mixture was concentrated to give the residue, which was dissolved with EtOAc (50 mL) and filtered. The filtrate was concentrated. The residue was purified by silica gel column chromatography eluting with PE/ EtOAc (10:1-6:1) to give the product, which was further washed with PE (10 mL) to obtain the title compound (3.43 g, 10.49 mmol, 60% yield) as a green solid. LCMS (Method d, Table 7) Rt 2.08 min; MS m/z 324.1 [M+H +].
Step 3: Synthesis of (2S,6aS,6bR7S,8aS,8bS,1OR, llaR,12aS,12bS)- l0-(4-((E) Aminostyry l)phenyl)-2,6b-difluoro- 7-hydroxy -8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d] [l,3]dioxolone E �CHO O BocHN "'= :::,._ ::,.. I MgS04 2] Trifluorometh anesulfonic acid (5.38 mL, 60.6 mmol) was added drop-wise to a 0 °C stirred s10n of (6S,8S,9R,1 OS,11S,13S, l 4S, l6R l7S)-6,9-difluoro-l l,16,17-trihydroxy- l7-(2- hydroxyacetyl)-10, 13-dimethyl-6,7,8,9,10,11,12,13,14,15 ,16, l7-dodecahydro-3H- cyclopenta[a]phenanthren one (Example 2, Step 5) (5.0 g, 12.12 mmol) and (E)-tert-butyl (3-(4 - for mylstyryl)ph enyl)carbamate (4.612 g, 12.12 mmol) in anhydrous MeCN (30 mL) and THF (30 mL) under nitrogen. The mixture was stirred at 0 °C for 1 h, then poured onto ice water (30 mL) and extracted withEtOAc (2 x 50 mL). The combined organic layers were washed with cooled water (30 mL), brine (30 mL), saturated NaHCO 3 (30 mL) and water (30 mL) again, and concentrated in vacuo affor ding a yellow solid. The crude material was purified by silica gel column tography (200-300 mesh), eluting with DCM/ MeOH (100%-40: 1) and then r purified by prep-HPLC to give the title compound (1.45 g, 2.328 mmol, 19% yield). LCMS (Method d, Table 7) R1 n; MS m/z +]. 1H NMR 618.3 [M+H (400 MHz, DMSO-d6) 8 7.61 (d, J 8.2 Hz, 2H), 7.43 (d, J 8.3 Hz, 2H), 7.28 (d, J 10.9 Hz, lH), 7.15 (d, J 16.4 Hz, lH), 7.03 (dd, J 15.5, 7.6 Hz, 2H), 6.75 (dd, J 8.0, 4.7 Hz, 2H), 6.50 (dd, J 7.9, 1.3 Hz, lH), 6.31 (dd, J= 10.1, 1.8 Hz, lH), 6.15 (s, lH), 5.79 5.46 (m, 3H), 5.13 (dd, J= 14.7, 8.7 Hz, 3H), 4.97 (d, J 5.1 Hz, lH), 4.55 (dd, J 19.5, 6.4 Hz, lH), 4.23 (dd, J 19.4, 5.5 Hz, 2H), 2.73 2.56 (m, lH), 2.40 2.21 (m, 2H), 2.15 2.02 (m, lH), 1.82 1.64 (m, 3H), 1.61 1.44 (m, 4H), 0.88 (s, 3H).
The minor acetal isomer, (2S,6aS,6bR,7S,8aS,8bS,10S,1 laR,12aS,12bS)- l0-(4-((E) aminostyryl)pheny1)-2,6b-difluoro-7 -hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[ 2', l':4,5]indeno[l,2-d] [l,3]dioxolone (0.30 g, 0.456 mmol, 4% yield), also was ed asa white solid. LCMS (Method d, Table 7) R1 1.51 min; MS m/z +]. 1H NMR (400 MHz, DMSO- d 618.3 [M+H 6) 8 7.57 (d, J 8.2 Hz, 2H), 7.27 (d, J 7.9 Hz, 3H), 7.18 6.97 (m, 3H), 6.75 (d, J 7.8 Hz, 2H), 6.50 (d, J 7.4 Hz, lH), 6.31 (dd, J 10.2, 1.6 Hz, lH), 6.15 (d, J 10.3 Hz, 2H), 5.78 5.67 (m, lH), 5.63 5.49 (m, 2H), 5.37 (d, J 7.0 Hz, lH), .07 (dd, J 12.0, 5.8 Hz, 3H), 4.33 4.15 (m, 2H), 4.06 (dd, J 19.2, 5.7 Hz, lH), 2.69 2.54 (m, lH), 2.36 2.08 (m, 3H), 1.94 1.60 (m, 4H), 1.50 (s, 3H), 0.90 (s, 3H).
Example 52: Synthesis of(6aR6bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((E) ty ryl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyl - l ,2,6a, ,8a,8b, l la, 12, 12a, 12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[ l,2-d][l,3]dioxolone o o Boc HN l� " � o H - � HO ,,. + #P "'- �: ... o� #P � OH HO� MgSO4 H2N H2N '<:: "': _, OH OH OH � �I Synthesized usmg the same procedure as Example 51 above. Major acetal isomer: (6aR6bS, 7S,8aS,8bS, 1 OR l laR,12aS,12bS)- l 0-( 4-((E)aminostyryl)phenyl)- 7-hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la, 12, 12a,12b-dodecahydro-4H- naphtho[2', l':4,5]indeno[ l,2-d][ oxolone. LCMS (Method d, Table 7) R1 1.48 min; MS m/z 582.3 [M+H+].1H NMR (400 MHz, DMSO-d 6) 8 0.88 (s, 3H), 1.00-1.09 (m, 2H), 1.40 (s, 3H), 1.63-1.79 (m, 5H), 2.04-2.15 (m, 2H), 2.32-2.34 (m, lH), 2.55-2.60 (m, lH), 4.20 (dd, J= 20.2 Hz, 5.0 Hz, lH), 4.31 (s, lH), 4.54 (dd, J= 19.2 Hz, 6.0 Hz, lH), 4.82 (s, lH), 4.94-4.95 (m, lH), 5.10-5.14 (m, 3H), 5.46 (s, lH), 5.95 (s, lH), 6.18 (d, J= 10.0 Hz, lH), 6.50 (d, J= 7.6 Hz, lH), .76 (m, 2H), 7.00-7.14 (m, 3H), 7.32 (d, J= 10.0 Hz, lH), 7.45 (d, J= 8.4 Hz, 2H), 7.59 (d, J= 8.0 Hz, 2H).
Minor acetal isomer : (6aR,6bS,7S,8aS,8bS, l0S, l S,12bS)(4-((E) aminostyryl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la, 12, 12a,12bdodecahydro-4H-naphtho [2', l':4,5]indeno[ l ,2-d][ l,3]dioxolone. LCMS (Method d, Table 7) R1 1.52 min; MS m/z +]. 1H NMR (400 MHz, DMSO-d 582.3 [M+H 6) 8 0.89 (s, 3H), 1.05-1.31 (m, 3H), 1.40 (s, 3H), .89 (m, 5H), 2.05-2.07 (m, 2H), 2.31-2.34 (m, lH), 2.54-2.59 (m, lH), 4.00-4.06 (m, lH), 4.23-4.31 (m, 2H), 4.80 (s, lH), 5.05-5.09 (m, 3H), 5.31-5.32 (m, lH), 5.95 (s, lH), 6.12 (s, lH), 6.18 (d, J= 9.6 Hz, lH), 6.49-6.50 (m, lH), 6.74-6.76 (m, 2H), 7.00-7.14 (m, 3H), 7.26 (d, J= 7.6 Hz, 2H), 7.32 (d, J= 10.0 Hz, lH), 7.57 (d, J= 7.6 Hz, 2H).
Example 53: Synthesis of(6aS,6bR,7S,8aS,8bS, l0R l aS,12bS)(4-((E)aminostyryl)phenyl)- oro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la,12,12a,12bdodecahydro-4H-naphtho [2', l ':4,5]indeno[1,2-d] [1,3]dioxolone o o Boc HN " --� O HO � ,,. F + #P "'- � F #P HO� MgSO4 H2N H2N '<:: "': _, OH OH OH �I � ,4, Synthesized usmg the same procedure as Example 51 above. Major acetal isomer: (6aS,6bR,7S,8aS,8bS, 1OR, l laR,12aS,12bS)-l0-(4-((E)aminostyryl)phenyl)-6b-fluorohydroxy-8b­ (2-hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-4H- naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone. LCMS (Method d, Table 7) R1 1.45 min; MS m/z 600.3 [M+H+]. 1H NMR (400 MHz, DMSO-d 6) 8 7.61 (d, J 8.1 Hz, 2H), 7.42 (d, J 8.1 Hz, 2H), 7.30 (d, J 10.1 Hz, lH), 7.15 (d, J 16.4 Hz, lH), 7.03 (dd, J 15.6, 7.6 Hz, 2H), 6.79 6.70 (m, 2H), 6.50 (d, J 7.7 Hz, lH), 6.25 (dd, J 10.1, 1.4 Hz, lH), 6.06 (s, lH), 5.54 5.43 (m, 2H), 5.13 (t, J 6.0 Hz, 3H), 4.95 (d, J 4.5 Hz, lH), 4.55 (dd, J 19.5, 6.4 Hz, lH), 4.22 (dd, J 19.3, 5.4 Hz, 2H), 2.62 (m, 2H), 2.42 2.02 (m, 3H), 1.92 1.80 (m, lH), 1.77 1.61 (m, 3H), 1.51 (s, 3H), 1.47 1.32 (m, lH), 0.89 (s, 3H).
Minor acetal isomer: (6aS,6bR,7S,8aS,8bS,l0S, l laR,12aS,12bS)( 4-((E) aminostyry1 )pheny fluoro- 7-hydroxy-Sb-(2-hydroxyacety1)-6a,8a-dimethyll ,2, 6a, 6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphth o[2', l':4,5]indeno[l,2-d][l,3]dioxolone.
LCMS (Method d, Table 7) R1 1.48 min; MS m/z +]. 1H NMR (400 MHz, 600.3 [M+H DMSO-d6) 8 7.57 (d, J 8.1 Hz, 2H), 7.28 (dd, J 12.2, 9.3 Hz, 3H), 7.18 6.97 (m, 3H), 6.75 (d, J 7.7 Hz, 2H), 6.50 (d, J 7.3 Hz, lH), 6.25 (dd, J 10.1, 1.2 Hz, lH), 6.13 (s, lH), 6.05 (s, lH), 5.46 (d, J 2.8 Hz, lH), 5.35 (d, J 6.9 Hz, lH), 5.06 (dd, J 14.0, 7.9 Hz, 3H), 4.24 (dd, J 19.3, 6.3 Hz, 2H), 4.05 (dd, J 19.1, 5.8 Hz, lH), 2.73 2.58 (m, lH), 2.47 2.30 (m, 2H), 2.09 (d, J 10.1 Hz, 2H), 1.85 (d, J 6.5 Hz, 2H), 1.78 1.65 (m, 2H), 1.50 (s, 4H), 0.90 (s, 3H).
Example 54: Synthesisof (2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l 0-(4-(3- aminophenethyl)phenyl)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b, 7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphth o[2', l':4,5]indeno[l,2-d][l,3]dioxolone Step 1: Synthesis oftert-Buty1 (E)-(3-(4-(hydroxymethyl)styryl)phenyl)carbamate CHO NaBH4 - � .A:,,... n MeOH � .A:,,... UOH BocHN "</ "</ BocHN "</ """' U U NaBH4 (0.936 g, 24.74 mmol) was added to a 0 °C solution of (E)-tert-butyl (3-(4- formylstyryl)phenyl)carbamate )Step 2, Example. 0 g, 12.37 mmol) in MeOH (60 mL) and THF (60 mL) and stirred at 0 °C for 1 h. The e was quenched with saturated aqueous NH4Cl (20 mL), concentrated to obtain a residue, which was ioned between EtOAc (100 mL) and water (100 mL).
The organic layer was trated under reduced re, and was purified by silica gel column chromatography, g with DCM/EtOAc (10:1-5:1) to give the title compound (3.23 g, 7.08 mmol, 57% yield) as a light red solid. LCMS (Method d, Table 7) R1 +]. 1.98 min; MS m/z 348.1 [M+Na Step 2: Synthesis oftert-Butyl (3-(4-(hydroxymethyl)phenethyl)phenyl)carbamate ,,,,__ .A:,,...UOH «OH EtOAc/THF _,,,,__ ,---._ BocHN """'"""' BocHN --...,,-"""' U U A suspension of Pd/C (0.657 g, 0.618 mmol) and (E)-tert-butyl (3-(4-(hydroxymethyl)styryl) phenyl)carbamate(pure) 7 (3.35 g, 10.29 mmol) in EtOAc (50 mL) and THF (50 mL) was treated with hydrogen balloon and stirred at 0 °C for 1.5 h, monitored by LCMS. The mixture was filtered. Additional Pd/C (0.657 g, 0.618 mmol) was added to the filtrate. The mixture was stirred for additional 1 h under an atmosphere of hydrogen and monitored by LCMS. The mixture was filtered and washed with EtOAc (15 mL). The filtrate was concentrated to give a residue, which was purified by silica gel column tography, g with PE/ EtOAc (10:1-2:1) to give the title compound (1.2 g, 3.49 mmol, 34% yield) as a white solid. LCMS (Method d, Table 7) R1 2.0 min; MS m/z +]. 350.0 [M+Na Step 3: Synthesis oftert-Butyl (3-(4-formylphenethyl)phenyl)carbamate OH Mn02 CHO DCM, 30 °C BQCHN I ---- BocHN�� ---::: A suspension of MnO2 (9.24 g, 106 mmol) and utyl (3-(4-(hydroxymethyl)phenethyl) phenyl)carbamate (2.9 g, 8.86 mmol) in DCM (40 mL) was treated with nitrogen balloon and stirred at 30 °C for 2 h, monitored by LCMS. Additional MnO2 (0.8 g, 9.2 mmol) was added to the above mixture, stirred at 30 °C for additional 1 h. The mixture was filtered and washed with DCM (20 mL). The filtrate was concentrated to obtain the title compound (2.9 g, 8.58 mmol, 97% yield) as a yellow solid. LCMS (Method d, Table 7) Rt 2.14 min; MS m/z 1H NMR (400 MHz, CDC1 226.0 [M-Boct. 3) 8 9.90 (s, lH), 7.71 (d, J 7.8 Hz, 2H), 7.25 (s, 2H), 7.19 6.97 (m, 3H), 6.73 (d, J 7.3 Hz, lH), 6.48 (s, lH), 2.98 2.87 (m, 2H), 2.86 2.78 (m, 2H), 1.44 (s, 9H).
Step 4: sis of (2S,6aS,6bR,7S,8aS,8bS,1 0R,l laR,12aS,12bS)(4-(3- Aminophenethyl)phenyl)-2,6b-difluoro hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b, 7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone "�;��OH -Boc - HN-�-1 �� ;�-�, = - - � e , ,u'-/� "¾ ~ D' • �' � OH � OH Trifluoromethanesulfonic acid (5.61 mL, 64.2 mmol) was added drop-wise to a stirred 0 °C suspension of tert-butyl formylphenethyl)phenyl)carbamate (4.18 g, 12.85 mmol) and (6S,8S,9R, l0S, l1S,13S,14S,16R,l7S)-6,9-difluoro- l l,16, l7-trihydroxy- hydroxyacetyl)-10,13- dimethyl-6,7,8,9,10,11,12,13,14,15,16, l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (5.3 g, 12.85 mmol) in anhydrous MeCN (30 mL) and THF (30 mL) under. The resulting mixture was stirred at 0 °C for 1 h, then poured onto ice water (20 mL) and extracted with EtOAc (2 x 25 mL). The combined c layers were washed with cooled water (20 mL), brine (10 mL), saturated aqueous NaHCO3 (20 mL) and water (20 mL), concentrated in vacuo affording a yellow solid. The crude material was purified by silica gel column chromatography (200-300 mesh), eluting with dichloromethane/ methanol (100%-40: 1) to obtain the product, which was r ed by PLC to give the title compound (2.21 g, 3.57 mmol, 28% yield) as a white solid. LCMS (Method d, Table 7) R1 1.75 min; MS m/z +]. 1H 619.8 [M+H NMR (400 MHz, DMSO-d6) 8 7.34 (d,J 8.1 Hz, 2H), 7.27 (t,J 8.0 Hz, 3H), 6.89 (t,J 7.7 Hz, lH), 6.43 (s, lH), 6.36 (d, J 7.9 Hz, 2H), 6.31 (dd, J 10.2, 1.8 Hz, lH), 6.14 (s, lH), 5.75 -5.56 (m, lH), .54 (d,J 2.9 Hz, lH), 5.46 (s, lH), 5.12 (t,J 6.0 Hz, lH), 4.95 (d,J 5.1 Hz, lH), 4.92 (s, 2H), 4.53 (dd, J 19.5, 6.4 Hz, lH), 4.21 (dd, J 19.4, 5.6 Hz, 2H), 2.83-2.79 (m, 2H), 2.73 -2.57 (m, 3H), 2.275- 2.25 (m, 2H), 2.08-2.04 (m, lH), 1.79 -1.62 (m, 3H), 1.67-1.50 (m, 4H), 0.87 (s, 3H).
The rumor acetal isomer, (2S,6aS,6bR,7S,8aS,8bS,1 0S,1 aS,12bS)- l 0-(4-(3- aminophenethyl)phenyl)-2,6b-difluoro-7 -hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxolone (0.45 g, 0.667 mmol, 5% yield) also was isolated as a white solid. LCMS (Method d, Table 7) R1 1.79 min; MS m/z +]. 1H NMR (400 MHz, DMSO-d 619.8 [M+H 6) 8 7.33-7.17 (m, 5H), 6.89 (t,J 7.7 Hz, lH), 6.46 - 6.27 (m, 4H), 6.12 (d,J 8.8 Hz, 2H), 5.75 - 5.55 (m, lH), 5.53 (s, lH), 5.34 (d, J 7.1 Hz, lH), 5.06 (t, J 5.9 Hz, lH), 4.92 (s, 2H), 4.31 -4.15 (m, 2H), 4.05 (dd,J 19.2, 5.6 Hz, lH), 2.83-2.79 (m, 2H), 2.72 -2.54 (m, 3H), 2.29 (s, lH), 2.21-2.13 (m, lH), 2.09-2.05 (m, lH), 1.93 -1.81 (m, lH), 1.79 - 1.60 (m, 3H), 1.50 (s, 3H), 0.88 (s, 3H).
Example 55: Synthesis of(6aR6bS,7S,8aS,8bS, l0R, l laR,12aS,12bS)(4-(3- aminophenethyl)phenyl)hydroxy-8b-(2-hydroxyacetyl )-6a,8a-dimethyll ,2,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxolone O BocHN C MgS04 8] Synthesized using the same procedure as Example 54 above. Major acetal isomer: LCMS d d, Table 7) Rt l.74 min ; MS m/z +]. 1H NMR (400 MHz, DMSO-d 583.8 [M+H 6) 8 7.34 (dd,J 16.7, 9.1 Hz, 3H), 7.24 (d,J 8.0 Hz, 2H), 6.89 (t,J 7.7 Hz, lH), 6.42 (s, lH), 6.36 (dd,J 7.7, 1.6 Hz, 2H), 6.17 (dd,J 10.1, 1.7 Hz, lH), 5.95 (s, lH), 5.41 (s, lH), 5.11 (t,J 5.9 Hz, lH), 4.93 (d,J .4 Hz, 3H), 4.81 (d,J 3.0 Hz, lH), 4.52 (dd,J 19.5, 6.4 Hz, lH), 4.30 (s, lH), 4.19 (dd,J 19.5, 5.6 Hz, lH), 2.87 -2.77 (m, 2H), 2.73 -2.64 (m, 2H), 2.62 -2.52 (m, lH), 2.32 (d,J 11.0 Hz, lH), 2.18 - 1.98 (m, 2H), 1.83 -1.58 (m, 5H), 1.40 (s, 3H), 1.24 -0.97 (m, 2H), 0.87 (s, 3H).
Minor acetal isomer, (6aR6bS,7S,8aS,8bS, l0S, l laR,12aS,12bS)(4-(3- henethyl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d] [l,3]dioxolone: LCMS (Method d, Table 7) Rt +]. 1H NMR (400 MHz, DMSO-d 1.77 min; MS m/z 583.9 [M+H 6) 8 7.32 (d,J 10.1 Hz, lH), 7.19 (q,J 8.2 Hz, 4H), 6.89 (t,J 7.7 Hz, lH), 6.44 - 6.29 (m, 3H), 6.17 (dd, J 10.1, 1.8 Hz, lH), 6.07 (s, lH), 5.95 (s, lH), 5.29 (d,J 6.9 Hz, lH), 5.03 (t,J 6.1 Hz, lH), 4.92 (s, 2H), 4.78 (d, J= 3.1 Hz, lH), 4.34 -4.19 (m, 2H), 4.02 (dd, J= 19.2, 5.9 Hz, lH), 2.81 (dd, J= 9.5, 6.1 Hz, 2H), 2.68 (dd, J 9.6, 6.0 Hz, 2H), 2.61 - 2.52 (m, lH), 2.32 (d, J 10.4 Hz, lH), 2.03 (d, J 7.8 Hz, 2H), 1.91 -1.67 (m, 5H), 1.39 (s, 3H), 1.27 -1.01 (m, 2H), 0.89 (s, 3H).
Example 56: Synthesis of (6aS,6bR,7S,8aS,8bS,l0R, l laR,12aS,12bS)(4-(3- henethyl)phenyl)-6b-fluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone O BocHN C MgS04 Synthesized using the same procedure as Example 54above. Major acetal isomer: LCMS (Method d, Table 7) Rt l.74 min; MS m/z +]. 1HNMR (400 MHz, DMSO-d 601.9 [M+H 6) 8 7.32 (t, J 7.2 Hz, 2H), 7.26 (t, J 8.0 Hz, 2H), 6.89 (t, J 7.7 Hz, lH), 6.43 (s, lH), 6.36 (d, J 7.7 Hz, 2H), 6.24 (dd, J 10.1, 1.7 Hz, lH), 6.05 (s, lH), 5.45 (s, 2H), 5.10 (t, J 5.9 Hz, lH), 4.97 -4.85 (m, 3H), 4.52 (dd, J 19.5, 6.4 Hz, lH), 4.20 (dd, J 19.2, 5.6 Hz, 2H), 2.85 -2.76 (m, 2H), 2.72 -2.54 (m, 3H), 2.36 (d, J 10.4 Hz, lH), 2.20-2.18 (m, lH), 2.04 (s, lH), 1.91 - 1.80 (m, lH), 1.73 -1.61 (m, 3H), 1.50 (s, 3H), 1.40-1.38 (m, lH), 0.87 (s, 3H). 1] Minor acetal isomer, (6aS,6bR,7S,8aS,8bS, 1OS, l laR,12aS,12bS)- l 0-( 4-(3- aminophenethyl)phenyl)-6b-fluorohydroxy-8b-(2-hydroxy acetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone: LCMS (Method d, Table 7) R1 1.77 min; MS m/z +]. 1HNMR (400 MHz, DMSO-d 601.9 [M+H 6) 8 7.35 -7.13 (m, 5H), 6.89 (t, J 7.7 Hz, lH), 6.41 (s, lH), 6.36 (d, J 7.6 Hz, 2H), 6.24 (dd, J 10.1, 1.7 Hz, lH), 6.06 (d, J 13.8 Hz, 2H), 5.44 (d, J 2.6 Hz, lH), 5.33 (d, J 7.0 Hz, lH), 5.04 (t, J 6.0 Hz, lH), 4.91 (s, 2H), 4.27-4.21 (m, 2H), 4.04 (dd, J 19.2, 5.9 Hz, lH), 2.85 -2.76 (m, 2H), 2.70-2.66 (m, 3H), 2.37-2.35 (m, 2H), 2.07-2.06 (m, 2H), 1.84 (d, J= 7.1 Hz, 2H), 1.71 (t, J= 10.3 Hz, 2H), 1.50 (s, 4H), 0.90 (s, 3H).
Example 57: Synthesis of (6aR,6bS,7S,8aS,8bS,l0R, llaR,12aS,12bS)(4-((3- Aminophenyl)amino)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a, 8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-4H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxolone Step 1: Synthesis of tert-Butyl (3-( (4-formyl )amino)phenyl)carbamate pBrPhCHO b, BINAP H BocHN v NH2 � Cs2C03, PhMe BocHN N yY V Y) A mixture tert-butyl (3-aminophenyl)carbamate (31.2 g, 150 mmol), 4-bromobenzaldehyde (33.3 g, 180 mmol), Pd(OAch (1.684 g, 7.50 mmol), BINAP ((RS)2,2'-bis(diphenylphosphino)- l,l'­ binaphthyl) (9.34 g, 15.00 mmol), Cs2CO3 (98 g, 300 mmol) was refluxed in toluene (300 mL) under nitrogen for 16 h. After cooling to room temperature, the e was partitioned between water and EtOAc. The organic layer was concentrated and purified by column tography g with PE:EtOAc (5: 1) to give the title compound (32.8 g, 105 mmol, 70% yield) as a yellow oil. LCMS (Methodj, Table 7) R1 1.94 min; MS m/z +]. 313 [M+H Step 2: Synthesis of (6aR,6bS,7S,8aS,8bS, l0R,l laR,12aS,12bS)(4-((3- Aminophenyl)amino)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2, 6a, 6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxolone �0 U BocHNv HO H CHO 0 OH TfOH MgS04 Trifluoromethanesulfonic acid (14.21 ml, 160 mmol) was added ise to a 0 °C suspension of (8S,9S,10R,11S,13S,14S,16R,l7S)-l l,16, l 7-trihydroxy-l7-(2-hydroxyacetyl)- 10,13- yl-6, 7,8,9, 10,11,12, 13,14,15,16, l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (12.05 g, 32.0 mmol) and tert-butyl (3-((4-form ylphenyl)amino)phenyl)carbamate (10 g, 32.0 mmol) in THF (50.00 ml) and MeCN (50 ml). The reaction mixture was stirred for additional 2 hours at the same temperature.
The mixture was diluted with EtOAc (200 mL), washed with water (100 mL), saturated NaHCO 3 solution (lx 100 mL), and brine (lxl00 mL), dried over Na2SO4, and concentrated in vacuo. The residue was purified by column chromatography (MeOH:DCM 1 :40), and the ing material was ed further by prep-HPLC to afford the title compound (1.729 g, 3.03 mmol, 10% yield) as a white solid. LCMS (Method k, Table 7) R1 1.50 min; MS m/z +]. 1H NMR (400 MHz, DMSO) 57 l[M+H 8 8.01 (s, lH), 7.33 (d, J 10.1 Hz, lH), 7.27 (d, J 8.5 Hz, 2H), 7.00 (d, J 8.5 Hz, 2H), 6.87 (t, J 7.9 Hz, lH), 6.36 (s, lH), 5.94 (s, lH), 5.32 (s, lH), 5.10 (s, lH), 5.02 4.87 (m, 3H), 4.80 (d, J 2.8 Hz, lH), 4.51 (d, J 16.4 Hz, lH), 4.31 (s, lH), 4.20 (d, J 17.8 Hz, lH), 2.62 2.52 (m, lH), 2.32 (d, J I 1.0 Hz, lH), 2.20 1.98 (m, 2H), 1.86 1.69 (m, 4H), 1.69 1.55 (m, lH), 1.41 (s,3H), 1.18 0.97 (m, 2H), 0.87 (s, 3H).
The rumor acetal isomer, (6aR,6bS,7S,8aS,8bS,10S, l laR,12aS,12bS)(4-((3- aminophenyl)amino)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2, 6a, 6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxolone (78 mg, 0.137 mmol, 0.4% yield) as a white solid. LCMS (Method k, Table 7) R1 1.53 min; MS m/z 57l[M+H+]. 1H NMR (400 MHz, DMSO) 8 8.00 (s, lH), 7.32 (d, J= 10.1 Hz, lH), 7.07 (d, J= 8.5 Hz, 2H), 6.96 (d, J 8.5 Hz, 2H), 6.87 (t, J 7.9 Hz, lH), 6.35 (s, lH), 6.24 (d, J 7.9 Hz, lH), 6.17 (d, J .0 Hz, lH), 6.10 (d, J 7.9 Hz, lH), 6.00 (s, lH), 5.95 (s, lH), 5.27 (d, J 7.0 Hz, lH), 5.02 (t, J 5.9 Hz, lH), 4.97 (s, 2H), 4.78 (d, J 2.7 Hz, lH), 4.30 (s, 2H), 4.03 (dd, J 19.1, 5.8 Hz, lH), 2.65 2.52 (m, lH), 2.32 (d, J= I0.2 Hz, lH), 2.14 - 1.95 (m, 2H), 1.89 - 1.63 (m, 5H), 1.39 (s, 3H), 1.28 - 1.11 (m, lH), 1.05 (d, J= 10.7 Hz, lH), 0.89 (s, 3H).
Example 58: Synthesis of(6aS,6bR,7S,8aS,8bS, l0R, l laR,12aS,12bS)(4-((3- Aminophenyl)amino)phenyl)-6b-fluoro- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a,6b, ,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxolone Synthesized using the same procedure as e 57 above. Major acetal isomer: LCMS (Method k, Table 7) Rt=l.49 min; MS m/z = 589 [M+H+]. 1H NMR (400 MHz, DMSO) 8 8.02 (s, lH), 7.30 (d, J= 10.1 Hz, lH), 7.23 (d, J= 8.5 Hz, 2H), 6.99 (d, J= 8.5 Hz, 2H), 6.87 (t, J= 7.9 Hz, lH), 6.36 (s, lH), 6.31 - 6.16 (m, 2H), 6.10 (d, J= 7.8 Hz, lH), 6.04 (s, lH), 5.45 (d, J= 2.6 Hz, lH), 5.35 (s, lH), .11 (t, J= 5.9 Hz, lH), 4.97 (s, 2H), 4.91 (d, J= 4.6 Hz, lH), 4.51 (dd, J= 19.5, 6.3 Hz, lH), 4.20 (dd, J = 19.2, 5.5 Hz, 2H), 2.74 - 2.58 (m, lH), 2.36 (d, J= 10.2 Hz, lH), 2.27 - 2.13 (m, lH), 2.06 (d, J= 9.5 Hz, 2H), 1.93 - 1.78 (m, lH), 1.78 - 1.57 (m, 3H), 1.51 (s, 3H), 1.42 (dd, J= 12.4, 4.5 Hz, lH), 0.87 (s, Minor acetal isomer, (6aS,6bR,7S,8aS,8bS,1 OS, l laR,12aS,12bS)- l 0-( 4-( (3- henyl)amino)phenyl)-6b-fluoro-7 -hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12, 12a, 12b-dodecahydro-4H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxolone : LCMS (Method k, Table 7) Rt=l.50 min; MS m/z =589 [M+H+]. 1H NMR (400 MHz, DMSO) 8 8.01 (s, lH), 7.29 (d, J= 10.1 Hz, lH), 7.08 (d, J= 8.5 Hz, 2H), 6.97 (d, J= 8.5 Hz, 2H), 6.87 (t, J= 7.9 Hz, lH), 6.35 (s, lH), 6.24 (d, J= 8.6 Hz, 2H), 6.10 (d, J= 7.7 Hz, lH), 6.02 (d, J= 18.4 Hz, 2H), 5.44 (s, lH), .30 (d, J= 7.1 Hz, lH), 4.97 (s, 3H), 4.30 (d, J= 19.1 Hz, lH), 4.19 (d, J= 9.2 Hz, lH), 4.05 (d, J= 19.1 Hz, lH), 2.78 - 2.56 (m, lH), 2.36 (d, J= 13.3 Hz, lH), 2.06 (d, J= 10.7 Hz, 2H), 1.83 (dd, J= 16.3, 10.0 Hz, 2H), 1.76 - 1.61 (m, 2H), 1.50 (s, 4H), 0.89 (s, 3H).
Example 59: Synthesis of(2S,6aS,6bR,7S,8aS,8bS,1 OR, l laR,12aS,12bS)- l 0-( 4-( (3- Aminophenyl)amino)phenyl)-2, 6b-difluorohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2,6a, 6b,7,8,8a,8b, l la,12, 12a, 12b-dodecahydro-4H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxolone Synthesized using the same ure as Example 57above. Major acetal isomer: LCMS (Method 1, Table 7) R1 1.62 min; MS m/z +]. 1H NMR (400 MHz, DMSO) 8 8.02 (s, lH), 607 [M+H 7.25 (dd, J 18.2, 9.4 Hz, 3H), 6.99 (d, J 8.6 Hz, 2H), 6.87 (t, J 7.9 Hz, lH), 6.36 (t, J 1.9 Hz, lH), 6.30 (dd, J 10.1, 1.8 Hz, lH), 6.25 -6.19 (m, lH), 6.14 (s, lH), 6.09 (dd, J 7.9, 1.3 Hz, lH), 5.77 - .55 (m, lH), 5.53 (d, J 2.8 Hz, lH), 5.35 (s, lH), 5.11 (t, J 6.0 Hz, lH), 4.96 (s, 2H), 4.92 (d, J 5.1 Hz, lH), 4.51 (dd, J 19.5, 6.4 Hz, lH), 4.21 (dd, J 19.3, 5.6 Hz, 2H), 2.76 -2.53 (m, lH), 2.28 (dd, J 12.6, 5.9 Hz, 2H), 2.06 (d, J 12.0 Hz, 3H), 1.70 (dt, J 20.2, 6.0 Hz, 3H), 1.60 - 1.40 (m, 4H), 0.86 (s, 3H).
Minor acetal isomer, (2S,6aS,6bR,7S,8aS,8bS,10S,1laR,12aS,12bS)- l 0-(4-((3- aminophenyl)amino)phenyl)-2, luo rohydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2, 6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[ 2', ]indeno[l,2-d][l,3]dioxolone : LCMS (Method 1, Table 7) R1 1.65 min; MS m/z +]. 1HNMR (400 MHz, DMSO) 8 8.01 (s, 607 [M+H lH), 7.27 (d, J 9.8 Hz, lH), 7.09 (d, J 7.9 Hz, 2H), 6.97 (d, J 8.0 Hz, 2H), 6.88 (t, J 7.7 Hz, lH), 6.41 -6.19 (m, 3H), 6.20 -6.07 (m, 2H), 6.03 (s, lH), 5.65 (d, J 46.4 Hz, lH), 5.52 (s, lH), 5.32 (d, J 6.8 Hz, lH), 5.06 (s, lH), 4.97 (s, 2H), 4.32 (dd, J 19.1, 5.3 Hz, lH), 4.19 (s, lH), 4.06 (dd, J 18.7, 4.8 Hz, lH), 2.59 (d, J 13.8 Hz, lH), 2.29 (s, lH), 2.17 (d, J 7.2 Hz, lH), 2.07 (s, lH), 1.87 (d, J 6.7 Hz, lH), 1.69 (dd, J 23.7, 12.4 Hz, 3H), 1.50 (s, 4H), 0.89 (s, 3H).
Example 60: Synthesis of (6aR,6bS,7S,8aS,8bS,l0R,llaR,12aS,12bS)(4-((3- Aminobenzyl)thio)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2, 6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-4H-naphtho[ 2', l':4,5]indeno[l,2-d][l,3]dioxolone Step 1: Synthesis 3-Nitrobenzyl)thio)ben zaldehyde p-FPhCHO CHO SH K2C03, DMSO � V 02N .)0 To a solution of (3-nitrophenyl)methanethiol (35 g, 282 mmol) and rob enzaldehyde (52.5 g, 310 mmol) in dry dimethyl sulfoxide (220 mL) was added potassium carbonate (78 g, 564 mmol).
The reaction mixture was heated to 100°C for 4 hours. One onal vial was set up as described above.
The two reactions were combined and diluted with water (2 L) and then extracted with EtOAc (3 x 600 mL). The combined organic layer was dried over Na2S04, and concentrated to give a residue, which was purified by column chromatography (eluted with PE/EtOAc 20/1 to 5/1) to give the title compound (62 g, 80% yield) as slight brown solid. 1HNMR (400 MHz, DMSO-d 6) 8 4.54 (s, 2 H) 7.51 (d, J 8.33 Hz, 2 H) 7.59 (s, 1 H) 7.77 (d, J 8.33 Hz, 2 H) 7.87 (d, J 7.89 Hz, 1 H) 8.05 - 8.10 (m, 1 H) 8.30 (s, 1 H) 9.87 (s, 1 H).
Step 2: Synthesis of (6aR,6bS, 7S,8aS,8bS,1 OR, l laR,12aS,12bS)Hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl-l 0-(4-((3-nitrobenzyl)thio)phenyl)-l,2,6a,6b, 7,8,8a,8b, l la,12,12a,12b- dodecahydro-4H-naphtho[2', I': 4,5]indeno[ 1,2-d][ 1,3 ]dioxol Trifluoromethanesulfonic acid (21.23 mL, 239 mmol) was added drop-wise to a 0 °C solution of (8S,9S,10R,l1S,13S,14S,16R,l 7S)-l l,16,17-trihydroxy- hydroxyacetyl)-10,13-dimethyl- 6,7,8,9,10, l l,12,13,14,15,16,17-dodecahydro-3H-cyclopenta[a]phenanthrenone (9 g, 23.91 mmol) and 4-((3-nitrobenzyl)thio)benzaldehyde (7.19 g, 26.3 mmol) in MeCN (500 mL). The reaction was stirred for I hour at 0 °C. Two additional vials were set up as described above. All three reactions were ed and poured into water (2 L). The resulting mixture was extracted with EtOAc (3 x 500 mL).
The combined organic layer was dried over Na2SO4, and concentrated to give a residue, which was purified by prep-HPLC to give the title compound (5.57 g, 16% yield) as white solid. LCMS (Method n Table 7): Rt 3.20 min; m/z +]. 1H NMR (400 MHz, DMSO-d 632.0 [M+H 6) 8 0.84 (s, 3 H) 0.93 -1.08 (m, 2 H) 1.37 (s, 3 H) 1.52 -1.76 (m, 5 H) 1.94 -2.15 (m, 2 H) 2.29 (br d, J ll.91 Hz, I H) 2.50 - 2.58 (m, I H) 4.15 (dd, J=l9.40, 5.51 Hz, I H) 4.27 (br d, J 2.87 Hz, I H) 4.39 (s, 2 H) 4.48 (dd, J=l9.40, 6.39 Hz, I H) 4.77 (d, J=3.09 Hz, I H) 4.89 (d, J=4.63 Hz, I H) 5.07 (t, J=5.95 Hz, I H) 5.38 (s, I H) .91 (s, I H) 6.15 (dd, J I0.14, 1.76 Hz, I H) 7.25 - 7.38 (m, 5 H) 7.55 (t, J=7.94 Hz, I H) 7.79 (d, J=7.72 Hz, I H) 8.04 (dd, J 8.16, 1.54 Hz, I H) 8.19 (d, J=l.76 Hz, I H) ] The minor acetal isomer, (6aR,6bS,7S,8aS,8bS,l0S,l laR,12aS,12bS)hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl- l (3-nitrobenzyl)thio)phenyl)- l,2,6a,6b,7,8,8a,8b,Ila, 12, 12a, ecahydro-4H-naphtho [2',l ':4,5]indeno[ [l,3]dioxolone (0.34 g, I% yield) also was obtained as a white solid. LCMS (Method n, Table 7): R1 3.28 min; MS m/z +]. 1H NMR (400 MHz, 631.8 [M+H DMSO-d6) 8 0.86 (s, 3 H) 0.98 - 1.05 (m, I H) 1.10 -1.21 (m, I H) 1.37 (s, 3 H) 1.66 -1.88 (m, 5 H) 1.94 -2.08 (m, 2 H) 2.29 (br dd, J=l3.23, 2.87 Hz, I H) 2.50 - 2.56 (m, I H) 3.99 (dd, J l9.18, 5.95 Hz, I H) 4.20 (dd, J=l9.07, 6.28 Hz, I H) 4.27 (br s, I H) 4.39 (s, 2 H) 4.77 (d, J=3.09 Hz, I H) 4.99 (s, I H) 5.26 (d, J 6.84 Hz, I H) 5.92 (s, I H) 6.04 (s, I H) 6.15 (dd, J=I0.03, 1.87 Hz, I H) 7.16 (d, J 8.38 Hz, 2 H) 7.26 -7.34 (m, 3 H) 7.55 (t, J=7.94 Hz, I H) 7.75 (d, J=7.72 Hz, I H) 8.05 (dd, J 8.16, 1.54 Hz, I H) 8.21 (t, J=l.76 Hz, I H).
Step 3: Synthesis of (6aR,6bS, 7S,8aS,8bS, I0R, l laR,12aS,12bS)(4-((3- Aminobenzyl)thio)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyll ,2, 6a,6b,7,8,8a,8b, Ila, 12,12a,12b-dodecahydro-4H-naphtho[2', l ':4,5]indeno[ l,2-d][l,3]dioxolone 0 0 01 '' Zn, AcOH 01 '' Lo .. •· ---- Lo .. •· 02N H2N I sD,,,· sD,,,· OH I OH v.0 v.0 A mixture of ( S, ,8bS, l0R, l laR,12aS,12bS droxy-8b-(2-hydroxyacetyl)- 6a,8a-dimethyl-l0-(4-((3-nitrobenzyl)thio)phenyl)- l,2,6a,6b,7,8,8a,8b, l la,12, 12a,12b-dodecahydro-4H­ naphtho[2', l':4,5]indeno[ l,2-d][ l,3]dioxolone (138 mg, 0.22 mmol), zinc (214 mg, 3.28 mmol), and acetic acid (0.4 ml, 6.99 mmol) in EtOAc (2 mL) was stirred at 40 °C for 2 hours. LCMS showed partial conversion to the desired aniline t. Added more zinc (71 mg, 1.09 mml) and stirred at 40 °C for an additional 2 hours. The solution was cooled to room temperature and partitioned between saturated aqueous NaHCO3 and EtOAc (3x). The ed organic layers were dried over Na2SO4 and purified by chromatography (silica gel) g with 0-5% MeOH in DCM to give the title compound (64 mg, 0.106 mmol, 49% yield). LCMS (Method r, Table 7) R1 0.77 min; MS m/z +]. 1H NMR (400 601.9 [M+H MHz, DMSO-d6) 8 7.35 (d, J 8.5 Hz, 2H), 7.32 7.24 (m, 3H), 6.89 (t, J 7.7 Hz, lH), 6.56 (q, J 2.3 Hz, lH), 6.47 (d, J= 7.4 Hz, lH), 6.40 (ddd, J= 7.6, 2.6, 1.4 Hz, lH), 6.15 (dd, J= 10.1, 1.8 Hz, lH), .95 5.89 (m, lH), 5.38 (s, lH), 5.03 (d, J 14.0 Hz, 3H), 4.90 (d, J 4.8 Hz, lH), 4.77 (d, J 3.5 Hz, lH), 4.54 4.44 (m, lH), 4.28 (s, lH), 4.16 (d, J 20.6 Hz, lH), 4.06 (d, J 2.3 Hz, 2H), 2.59 2.50 (m, lH), 2.30 (d, J 11.5 Hz, lH), 2.14 2.03 (m, lH), 1.97 (s, 2H), 1.88 1.67 (m, 4H), 1.63 (td, J 11.9, .4, 5.1 Hz, lH), 1.37 (d, J 1.9 Hz, 3H), 1.10 0.92 (m, 2H), 0.84 (s, 3H).
Example 61: Synthesis of (6aR,6bS,7S,8aS,8bS, l0R, llaR,12aS,12bS)(4-((2-Aminopyridin yl)methyl)phenyl)- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la,12, 12a,12bdodecahydro-4H-naphtho [2',l ':4,5]indeno[1,2-d][ 1,3]dioxolone Step 1: Synthesis of ( 6aR,6bS, 7S,8aS,8bS, lOR, llaR12aS,12bS)-l0-(4- (Bromomethyl)phenyl)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl -6a,6b,7,8,8a,8b, l la,12,12a,12bdecahydro-1H-naphtho [2', l ':4,5]indeno[1,2-d][ l,3]dioxol-4(2H)-one 0 0 p BrCH 2PhCHO TfOH, MgS04 ' HO''' 0'' �,,..L-d OH OH 4-(Bromomethyl)benzaldehyde (0.539 g, 2.71 mmol) was added to a 0 °C suspension of (8S,9S,10R l lS,13S,14S,16R,17S)- l 7-trihydroxy(2-hydroxyacetyl)-10,13-dimethyl - 6,7,8,9,10, l l,12,13,14,15,16,l7-dodecahydro-3H-cyclopenta[a]phenanthrenone (1.0738 g, 2.85 mmol), 4-(bromomethyl)benzaldehyde (0.539 g, 2.71 mmol), and MgSO4 (1.33 g, 11.05 mmol) in MeCN (18 ml).
Trifluoromethanesulfonic acid (2.0 g, 13.5 mmol) was added in a drop-wise manner, so as to maintain a temperature of less than 7 °C. The on was stirred for 4 min, whereupon it was ed by addition of saturated aqueous NaHCO 3 (20 mL) and extracted with EtOAc (60 mL). The combined organics were washed with brine (10 mL) and solvent was removed under reduced pressure. Purification by tography (silica, 40 g) eluting with a gradient of 0-5% MeOH/DCM gave the title compound ( 1.59 g, 2.85 mmol, 100% yield) as an ite foam (9:1 mixture of acetal diastereomers. terization is provided for the major acetal isomer: LCMS (Method r, Table 7) R1 1.04 min; MS m/z 557.2, 559.2 [M+H]. lH NMR (501 MHz, DMSO-d6) d 7.44 (s, 4H), 7.30 (dd, J= 10.1, 2.2 Hz, lH), 6.15 (ddd, J= .1, 4.8, 1.9 Hz, lH), 5.91 (t, J 1.7 Hz, lH), 5.43 (s, lH), 5.07 (s, lH), 4.93 (d, J 5.1 Hz, lH), 4.77 (dd, J 3.6, 0.9 Hz, lH), 4.67 (s, 2H), 4.51 (dd, J 19.4, 4.1 Hz, lH), 4.31 4.26 (m, lH), 4.17 (d, J 19.5 Hz, lH), 2.58 2.49 (m, lH), 2.30 (dd, J 12.9, 4.7 Hz, lH), 2.16 2.05 (m, lH), 1.99 (d, J 23.9 Hz, lH), 1.89 1.71 (m, 2H), 1.75 1.65 (m, lH), 1.67 1.57 (m, lH), 1.38 (s, 3H), 1.11 0.91 (m, 2H), 0.85 (s, 3H).
Step 2: Synthesis of (6aR,6bS,7S,8aS,8bS,1OR, l laR,12aS,12bS)- l 0-( 4-((2-Aminopyridin yl)methyl)phenyl)- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl- l,2,6a,6b,7,8,8a,8b, l la,12,12a,12bdodecahydro-4H-naphtho [2', l ':4,5]indeno[1,2-d][1,3]dioxolone 0 ¾ 0 Pd(dppf)Cl2 o••· K2C03 '--o '' + H2N � l,o yy l .ro· ,' N✓N ° Br (�{- ,CJ)''' � OH H2N OH A 20 mL vial was charged with bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4- (bromomethyl)phenyl)- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl-6a,6b,7,8,8a,8b, l la,12,12a,12bdecahydro-1H-naphtho [2', l ':4,5]indeno[ l,2-d][ l,3]dioxol-4(2H)-one (0.100 g, 0.179 mmol), ,5,5- tetramethyl-1,3,2- dioxaborolanyl)pyridinamine (0.039 g, 0.179 mmol), and K2C03 (0.099 g, 0.718 mmol) in degassed dioxane (2.0 ml)/water (0.200 mL) solution. The suspension was evacuated and back filled with dry N2 (3X). Pd(dppf)Ch (0.012 g, 0.016 mmol) was added and the vial was once again evacuated and back filled with dry N2. The reaction mixture was heated to 90°C. After 1.5 hours the starting material was consumed. The reaction was allowed to cool to room temperature, diluted with EtOAc (20 mL) and washed with water (25 mL) then brine (25 mL), dried over MgSO4, and solvent was removed under reduced pressure. Purification by chromatography (silica, 40 g) eluting with a gradient of 0-10% MeOH/CH2Ch gave a light tan solid. Further purification by reverse phase prep HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 50 mm).A gradient of MeCN (A) and 0.1% TFA in water (B)was used, at a flow rate of80 mL/min (0-5.0 min 15% A, 5.0-20 min linear nt 15-85% A, 20-25 min hold). Combined fractions were frozen and lized to give the title compound (27 mg, 0.047 mmol, 26% yield) as a white solid. LCMS (Method r, Table 7) R1 +]. 0.90 min; MS m/z 571.3 [M+H lH NMR (501 MHz, DMSO-d6) 8 7.93 (s, 2H), 7.82 (d, J 6.6 Hz, lH), 7.44 (d, J 8.1 Hz, 2H), 7.39 - 7.22 (m, 3H), 6.73 (d, J 8.1 Hz, lH), 6.69 (s, lH), 6.24 - 6.09 (m, lH), 5.93 (s, lH), 5.44 (s, lH), 4.94 (d, J= 5.1 Hz, lH), 4.80 (s, lH), 4.50 (d, J= 19.4 Hz, lH), 4.30 (s, lH), 4.19 (d, J= 19.4 Hz, lH), 3.99 (s, 2H), 2.61 - 2.51 (m, lH), 2.35 - 2.27 (m, lH), 2.19 -2.08 (m, lH), 2.08 -1.99 (m, lH), 1.82 -1.59 (m, 5H), 1.40 (s, 3H), 1.02 (ddd,J 27.9 , 11.7 , 3.2 Hz, 2H), 0.87 (s, 3H).
The following examples were synthesized using the same ure as Example 61 (above).
Table 8.
Example Structure and Name LCMS and 1H NMR 0 LCMS (Method r, Table 7) R1 0.87 min; MS m/z 571.4 [M+H]. 1HNMR(500 MHz, DMSO-d6) 8 7.84 (d, J= 2.1 Hz, 2H), 7.75 (dd,J 9.1, 2.1 Hz, lH), 7.44 OH - 7.37 (m, 2H), 7.31 (d,J 10.1 Hz, lH), 7.26 (6aR,6bS,7S,8aS,8bS, l0R, l laR,12aS,12b (d,J 8.2 Hz, 2H), 6.90 (d, J 9.0 Hz, lH), S)(4-((6 pyridin 6.17 (dd,J 10.1, l.9 Hz, lH), 5.93 (t,J= 1.6 62 yl)methyl)phenyl)hydroxy-8b-(2- Hz, lH), 5.42 (s, lH), 5.10 (s, lH), 4.92 (d,J hydroxyacetyl)-6a,8a- dimethyl- 4.9 Hz, lH), 4.80 (d,J 3.4 Hz, lH), 4.50 (d, J l ,2,6a, 6b,7,8,8a,8b, l 12a,12b­ 19.4 Hz, lH), 4.29 (s, lH), 4.17 (d, J 19.5 dodecahydro-4H- Hz, lH), 3.84 (s, 2H), 2.61 -2.52 (m, 2H), 2.31 naphtho[2 ', l ':4,5]indeno[l,2- (d,J 12.3 Hz, lH), 2.13 (d, J 10.9 Hz, lH), d][l,3]dioxolone 2.08 -1.98 (m, lH), 1.81 - 1.58 (m, 5H), 1.40 (s, 3H), 1.00 (ddd,J 32.4 , 11.7, 4.1 Hz, 2H), 0.86 (s, 3H). 0 LCMS (Method r, Table 7) R1 0.87 min; MS m/z 571.4 [M+H]. 400 MHz, DMSO-d6)87.88 (s, lH), 2.2 Hz, lH), 7.36 (d, J 8.1 Hz, OH 7.81 (d,J bS,7S,8aS,8bS, l0R, l laR,12aS,12b 2H), 7.32 - 7.18 (m, 3H), 6.10 (dd,J 10.1, 1.8 63 S)(4-((5 -aminopyridin Hz, lH), 5.87 (s, lH), 5.36 (s, lH), 4.86 (d,J yl)methyl)phenyl)hydroxy-8b-(2- 4.8 Hz, lH), 4.74 (s, lH), 4.43 (d,J 19.5 Hz, hydroxyacetyl)-6a,8a- dimethyl- lH), 4.23 (s, lH), 4.11 (d,J 19.4 Hz, lH), l ,2,6a, 6b,7,8,8a,8b, l la,12,12a,12b­ 3.93 (s, 2H), 2.54 -2.45 (m, lH), 2.31 - 2.17 (m, dodecahydro-4H- lH), 2.14 -1.90 (m, 2H), 1.76 -1.50 (m, 5H), naphtho[2 ', l ':4,5]indeno[l,2- 1.33 (s, 3H), 1.05 - 0.85 (m, 2H), 0.80 (s, 3H) d][l,3]dioxolone 0 LCMS (Method r, Table 7) R1 0.86 min; MS m/z 571.3 [M+H]. 1HNMR(501 MHz, DMSO-d6) 8 7.89 (s, 2H), ''' 0.L_ ,,· � () 7.82 (d,J 6.2 Hz, lH), 7.58 (d, J 6.1 Hz, 64 OH �� lH), 7.36 (d,J 8.1 Hz, 2H), 7.23 (dd,J 20.2, 9.1 Hz, 3H), 6.85 - 6.71 (m, lH), 6.10 (d,J (6aR,6bS,7S,8aS,8bS, l0R, l laR,12aS,12b 11.9 Hz, lH), 5.86 (s, lH), 5.36 (s, lH), 4.87 (d, S)(4-((2 -aminopyridin J 5.0 Hz, lH), 4.75 (s, lH), 4.44 (d, J 19.4 yl)methyl)phenyl)hydroxy-8b-(2- Hz, lH), 4.23 (s, lH), 4.12 (d,J 19.4 Hz, lH), hydroxyacetyl)-6a,8a- dimethyl- l,2,6a,6b, 7,8,8a,8b, lla,12,12a,12b­ 3.87 (s, 2H), 2.55 - 2.45 (m, lH), 2.24 (d, J= dodecahydro-4H- 11.0 Hz, lH), 2.05 (d, J 22.0 Hz, lH), 2.01 - naphtho[2 ', l ':4,5]indeno[l,2- 1.91 (m, lH), 1.77 - 1.49 (m, 5H), 1.33 (s, 3H), ]dioxolone 1.04 - 0.87 (m, 2H), 0.80 (s, 3H).
E LCMS (Method r, Table 7) R1 0.85 min; MS m/z 607.4 [M+H].
.L_ 1HNMR (500 MHz, DMSO-d6) 8 7.80 (d, J= 6.5Hz, lH), 7.59 (s, 2H), 7.41 (d,J 8.2 Hz, H2N OH 2H), 7.31 (d, J= 7.9 Hz, 2H), 7.27 (d, J= 10.8 (2S,6aS,6bR,7S,8aS,8bS,1 OR l laR,12aS, 12bS)(4-((2-aminopyridin Hz, lH), 6.69 (d, J 6.8 Hz, lH), 6.64 (s, lH), yl)methyl)phenyl)-2,6b-difluoro 6.30 (dd,J 10.2, l.9 Hz, lH), 6.13 (s, lH), hydroxy-8b-(2-hydroxyacetyl)-6a,8a­ 5.55 (d, J= 5.8 Hz, lH), 5.49 (s, lH), 5.13 (s, lH), 4.96 (d, J dimethyl- 4.5 Hz, lH), 4.26 - 4.13 (m, l,2,6a,6b, 7,8,8a,8b, lla,12,12a,12b­ 2H), 3.95 (s, 2H), 2.62 - 2.53 (m, lH), 2.35 - 2.28 (m, lH), 2.28 - 2.18 (m, lH), 2.09 - 1.99 dodecahydro-4H- (m, lH), 1.77 -1.66 (m, 3H), 1.50 (s, 4H), 0.87 naphtho[2 ', l indeno[l,2- (s, 3H). d][l,3]dioxolone F LCMS (Method r, Table 7) R1 0.85 min ; 0 MS m/z 607.4 [M+H]. o•·· 1HNMR (400 MHz, DMSO-d6) 8 7.76 (s, lH), H2N ,,··L_cJ_. � 7.65 (d, J 9.5 Hz, lH), 7.59 (s, lH), 7.31 (d, J OH 8.2 Hz, 2H), 7.25 - 7.15 (m, 3H), 6.79 (d, J= (2S,6aS,6bR,7S,8aS,8bS,1 OR l laR,12aS, 9.2 Hz, lH), 6.23 (d, J 10.2 Hz, lH), 6.06 (s, 66 12bS)(4-((6-aminopyridin lH), 5.69 - 5.49 (m, lH), 5.46 (d, J 4.0 Hz, yl)methyl)phenyl)-2,6b-difluoro lH), 5.40 (s, lH), 5.03 (s, lH), 4.88 (d, J 4.2 hydroxy-8b-(2-hydroxyacetyl)-6a,8a­ Hz, lH), 4.44 (d, J 19.4 Hz, lH), 4.21 - 4.05 dimethyl- (m, 2H), 3.76 (s, 2H), 2.67 - 2.51 (m, lH), 2.32 - l,2,6a,6b, 7,8,8a,8b, lla,12,12a,12b­ 2.09 (m, 2H), 1.96 (d,J 13.1 Hz, lH), 1.71 - dodecahydro-4H- 1.57 (m, 3H), 1.50 -1.35 (m, 4H), 0.80 (s, 3H). naphtho[2 ', l ':4,5]indeno[l,2- d][l,3]dioxolone Example 67: Synthesis of 1-(3-(4-((6aR,6bS,7S,8aS,8bS,lOR, l laR, 12aS,12bS)Hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la, 12, 12a,12b-dodecahydro-1H­ o[2',l':4,5]indeno[1,2-d][1,3]dioxol-1O-yl)benzyl)phenyl)-lH-pyrrole-2,5-dione OH(Yy\"" 0 \ � 0 ,,::::::-.... .',·-y , .,,o N 0 o==<::{" 3] Step 1: sis of (Z)((3-(4-((6aR6bS,7S,8aS,8bS, l0R,l laR,12aS,12bS)Hydroxy- 8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H­ naphtho[2', l ': 4,5]indeno[ 1,2-d] [ l,3]dioxol-l0-yl)benzyl)phenyl)amino)oxobutenoic acid OH(fy\ "" OH(fy\ "" 0 \ .' ' I .0 o I '° 0 __,,:::,.. .' ' I0 __,,:::,.,. .,,0 .. ,o oy·· NH �C02H 0 0 Maleic anhydride (46.5 mg, 0.474 mmol) was added to a room temperature solution of (6aR6bS,7S,8aS,8bS,1OR, l laR,12aS,12bS)- (3-aminobenzyl)phenyl)hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyl-6a,6b,7,8,8a,8b,l la,12,12a,12b-decahydro-1H- naphtho[2', l ':4,5]indeno[ l,2-d][l,3]dioxol-4(2H)-one (239 mg, 0.420 mmol) in THF (3.0 mL). After 75 min, solvent was removed under reduced pressure to give the title compound as an off-white foam. This was used without further purification in the next step (100% yield was d). LCMS (Method o, Table 7) R1 0.86 min; MS m/z +]. 1H NMR (400 MHz, DMSO-d6) 8 12.99 (s, lH), 10.29 (s, lH), 668.5 [M+H 7.45 7.38 (m, 2H), 7.38 7.31 (m, 2H), 7.27 (d, J 10.1 Hz, lH), 7.24 7.15 (m, 3H), 6.92 (dt, J 7.8, 1.3 Hz, lH), 6.38 (d, J 12.1 Hz, lH), 6.25 (d, J 12.0 Hz, lH), 6.12 (dd, J 10.1, 1.9 Hz, lH), 5.89 (d, J 1.5 Hz, lH), 5.36 (s, lH), 5.03 (s, lH), 4.88 (d, J 5.1 Hz, lH), 4.73 (d, J 3.3 Hz, lH), 4.46 (d, J 19.4 Hz, lH), 4.26 (p, J 3.2 Hz, lH), 4.14 (d, J 19.4 Hz, lH), 3.87 (s, 2H), 2.52 (dd, J 13.6, 5.3 Hz, lH), 2.32 2.23 (m, lH), 2.07 (tt, J 10.8, 6.2 Hz, lH), 2.02 1.94 (m, lH), 1.84 1.51 (m, 5H), 1.36 (s, 3H), 1.09 0.93 (m, 2H), 0.82 (s, 3H).
Step 2: sis of 1-(3-(4-((6aR6bS,7S,8aS,8bS,1 OR l laR,12aS, 7-Hydroxy-8b-(2- hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H- naphtho[2', l ':4,5]indeno[1,2-d] [1,3]dioxol-1 0-yl)benzyl)phenyl)- lH-pyrrole-2,5-dione OH((y\ ""' o I '° ZnBr2, HMDS ,,:::,.,.
THF .::- 01·' N 0 .,,0 0� Bis(trimethylsilyl)amine (HMDS) (63.4 µL, 0.306 mmol) was added to a solution of zinc bromide (75.0 mg, 0.333 mmol) and (Z)((3-(4-((6aR,6bS,7S,8aS,8bS, l0R,l laR12aS,12bS) hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro- 1H-naphtho[2', l':4,5]indeno[ l,2-d][l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxobutenoic acid (171 mg, 0.256 mmol) in tetrahydrofuran (2.0 mL). The mixture was heated to 50 °C for 2.5 h. LCMS indicated incomplete conversion, so another aliquot of bis(trimethy lsilyl)amine (HMDS) (63.4 µL, 0.306 mmol) was added. The reaction was complete after an additional 90 min at 50 °C. The mixture was cooled to room temperature, d with EtOAc (20 mL), then washed tially with 1 N aqueous HCl (2 x mL), saturated aqueous NaHCO 3 (10 mL), brine (10 mL), dried over Na2SO4, and solvent was removed under reduced pressure. Purification by chromatography a, 12 g) eluting with a gradient of0-10% MeOH/DCM gave the title compound (82.6 mg, 0.127 mmol, 50% yield) as an off-white solid. LCMS (Method r, Table 7) R1 1.02 min; MS m/z 650.5 [M+H+]. 1H NMR (400 MHz, DMSO-d6) 8 7.27 (dt, J 7.8, 3.7 Hz, 3H), 7.19 (d, J 10.1 Hz, lH), 7.17 7.06 (m, 4H), 7.06 7.01 (m, 3H), 6.04 (dd, J 10.1, 1.9 Hz, lH), 5.81 (t, J 1.5 Hz, lH), 5.64 (s, lH), 5.29 (s, lH), 4.95 (t, J 5.9 Hz, lH), 4.80 (d, J 5.1 Hz, lH), 4.65 (d, J 3.2 Hz, lH), 4.38 (dd, J 19.4, 6.4 Hz, lH), 4.18 (t, J 3.4 Hz, lH), 4.06 (dd, J 19.5, .7 Hz, lH), 3.86 (s, 2H), 2.45 (dd, J 13.5, 5.4 Hz, lH), 2.30 2.11 (m, lH), 2.11 1.81 (m, lH), 1.76 1.44 (m, 4H), 1.28 (s, 3H), 1.02 0.83 (m, 2H), 0.75 (s, 3H).
Example 68: Synthesis of 2-((6aR,6bS,7S,8aS,8bS,1 OR, l aS,12bS)- l 0-(4-(3-(2,5-Dioxo-2,5- dihydro- lH-pyrrol- l -yl)benzyl)phenyl)hydroxy-6a,8a-dimethyloxo- 2,4,6a,6b, 7,8,8a,8b, l la, 12, 12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol-8b-yl) oxoethyl dihydrogen phosphate OH((y\ °""' o I � 0 ,,:::,.... phoryl chloride -' - (.. ,o o==<:;fN 0 THF 7] Diphosphoryl chloride (158 mg, 0.609 mmol) was added ise to a -51 °C solution of 1- (3-(4-((6aR,6bS,7S,8aS,8bS,1 OR, l laR,12aS,12bS)hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyl oxo-2,4,6a,6b, 7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol- l 0- yl)benzyl)phenyl)- lH-pyrrole-2,5-dione (82 mg, 0.126 mmol) in ydrofuran (0.5 mL). The reaction was slowly warmed to -10 °C over an hour then ed with water at -5°C. The mixture was treated with a saturated aqueous solution ofNaHCO 3 to give a solution with a pH of 8. Treatment with EtOAc (5 mL) gave a milky emulsion. Adjusting the pH to 1 by addition of 1 N aqueous HCl improved the emulsion. Extracted with EtOAc (4 x 5 mL), then washed the combined organics with brine (5 mL), dried (Na 2SO4), and removed solvent under reduced pressure. The product was purified by reverse phase prep HPLC on a Phenomenex Cl8(2) 10 micron column (250 x 50 mm). A gradient ofMeCN (A) and 0.1 % TFA in water (B) was used, at a flow rate of90 mL/min (0-5.0 min 15% A, 5.0-20.0 min linear gradient -95% A). ed fractions were frozen and lyophilized to give the title compound (3.6 mg, 4.93 mmol, 4% yield) as a white solid. LCMS (Method r, Table 7) R1 0.95 min; MS m/z 1H 730.5 [M+H+].
NMR (501 MHz, DMSO-d6) 8 7.38 (dt, J 7.8, 3.7 Hz, 3H), 7.31 (d, J 10.1 Hz, lH), 7.29 7.26 (m, 2H), 7.25 7.22 (m, lH), 7.19 (t, J 1.9 Hz, lH), 7.17 7.12 (m, 3H), 6.16 (dd, J 10.1, 1.9 Hz, lH), 5.93 (t, J 1.6 Hz, lH), 5.48 (s, lH), 4.96 4.86 (m, 2H), 4.84 (s, lH), 4.56 (dd, J 18.1, 8.1 Hz, lH), 4.30 (q, J 3.3 Hz, lH), 3.97 (s, 2H), 2.59 2.52 (m, lH), 2.31 (d, J 12.0 Hz, lH), 2.17 2.07 (m, lH), 2.05 1.98 (m, lH), 1.85 1.56 (m, 5H), 1.39 (s, 3H), 1.03 (ddd, J 18.5, 11.8, 4.1 Hz, 2H), 0.88 (s, 3H).
Example 69: Synthesis of 2-((6aR6bS,7S,8aS,8bS, l0R,l laR,12aS,12bS)(4-((3-((S)((S)(2- (2,5-Dioxo-2,5-dihydro-lH-pyrrol-l-yl)acetamido)propanamido)propanamido)phenoxy)methyl)phenyl)- 7-hydroxy-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H- naphtho[2', l indeno[l,2-d][l,3]dioxol-8b-yl)oxoethyl dihydrogen phosphate }- ,j( !Jl ,\, l oJ)' la y-y 0 � 0 = H O 0 HO,p 0 HO ''ci Step 1: Synthesis of utyl ((S)- l-(((S)((3-((4- ((6aR6bS,7S,8aS,8bS,1 OR, l laR, 12aS,12bS)-8b-(2-((di-tert-butoxyphosphoryl)oxy)acetyl)hydroxy- 6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2- d] [l,3]dioxol-l0-yl)benzyl)oxy)phenyl)amino)oxopropanyl)amino)oxopropanyl)carbamate 0 0 H H (tBu0)2PNEt2 tetrazole, then H2O2 0' o "', )--o BocHNJ �� ro � O N BocHN H yY � � O 0 OH � OP(O)(OtBu), Di-tert-butyl N,N-diethylphosphoramidite (0.226 ml, 0.811 mmol) was added to a room ature solution of tert-butyl ((S)- l-(((S)- l-((3-((4-((6aR,6bS,7S,8aS,8bS,l0R,l laR,12aS,12bS) hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7 ,8,8a,8b, l la, 12, 12a,12b-dodecahydro- 1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol-l0-yl)benzyl)oxy)phenyl)amino)oxopropan yl)amino)-l-oxopropanyl)carbamate , 0.559 mmol) and lH-tetrazole (0.45 Min MeCN, 4.97 ml, 2.237 mmol) in yl acetamide (2 ml). Additional di-tert-butyl N,N-diethylphosphoramidite (0.2 mL) was charged after 4.5 hours and stirring was continued overnight. The reaction was cooled to 0°C, whereupon a 30% solution of hydrogen peroxide in water (0.17 mL, 1.67 mmol) was added drop-wise.
Oxidation to the ate was complete within 1.5. The reaction was cooled to 0°C, and the reaction was quenched by addition of a IM aq. solution ofNa2S203 (8 mL). The mixture was extracted with EtOAc (2 x 30 mL), the combined organic layers were washed with brine, dried over Na2S04, and solvent removed under reduced pressure. Purification by chromatography (silica) using 100% EtOAc as eluent provided the title compound (366mg, 0.359 mmol, 64% yield) as white solid. LCMS (Method r, Table 7) R1 1.08 min; MS m/z +]. 1020.5 [M+H Step 2: Synthesis of2-((6aR6bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((3-((S)((S) Aminopropanamido namido )phenoxy)methyl)phenyl)hydroxy-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxol-8b-yl) oxoethyl dihydrogen phosphate o --s,,... L-o N O H, ��\� � OP(O)(OH)z TFA (0.95 mL) was added to a room temperature solution oftert-butyl ((S)- l-(((S)((3-((4- ((6aR6bS, 7S,8aS,8bS,1OR, l laR,12aS,12bS)-8b-(2-((di-tert-butoxyphosphoryl)oxy)acetyl)hydroxy- 6a,8a-dimethyloxo-2,4,6a,6b, 7,8,8a,8b,l la,12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2- d] [l,3]dioxol-l0-yl)benzyl)oxy)phenyl)amino)oxopropanyl)amino)oxopropanyl)carbamate (364 mg, 0.357 mmol) in DCM (2 mL). The reaction was complete within 2 h, whereupon t was removed under reduced pressure. The title nd was obtained as a foamy light yellow solid and was used without further purification. LCMS (Method r, Table 7) major acetal isomer: R1 0.77 min; MS m/z 808.3 [M+H+], minor acetal isomer: R1 0.79 min; MS m/z +]. 808.3 [M+H Step 3: sis of2-((6aR6bS,7S,8aS,8bS,l0R,l laR,12aS,12bS)(4-((3-((S)((S) (2-(2,5-Dioxo-2,5-dihydro-lH-pyrrol-lyl )acetamido)propanamido)propanamido)phenoxy)methyl)phenyl)hydroxy-6a,8a-dimethyloxo- 2,4,6a,6b, 7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxol-8b-yl) oxoethyl dihydrogen ate 0 0 H H & & 0'' o•·· o --,,)--o o o ,,,.Lo 1 Pr2NEt H 1 H __,.Q H,N O N N O & ""° O!B,), }- N"j( 0f n o - o OP(O)(OH), 4o Y")' � N, N-Diisopropylethylamine (0.37 mL, 2.12 mmol) and maleimidoacetic acid N­ hydroxysuccinimide ester (89 mg, 0.353 mmol) were added sequentially to a room temperature solution of 2-((6aR,6bS,7S,8aS,8bS, lOR, l laR12aS,12bS)-l 0-( 4-((3-((S)((S) aminopropanamido)propanamido)phenoxy)methyl)pheny1)hydroxy-6a,8a-dimethyloxo- 2,4,6a,6b,7,8,8a,8b,l la,12,12a,12b-dodecahydro-1H-naphtho[ 2', l ':4,5]indeno[l,2-d][l,3]dioxol-8b-yl) oxoethyl dihydrogen phosphate (285mg, 0.353 mmol) in dimethyl formamide (1.5 mL) and was stirred ght. The reaction e was diluted with DMSO and was purified by preparative reverse phase HPLC on a enex Cl8(2) 10 micron column (250x 50 mm). A gradient ofMeCN (A) and 0.1% TFA in water (B) was used, at a flow rate of 30mL/min (0-3.0 min 15% A, 3.0-19.0 min linear gradient -60% A, then 19.0-23.0 min linear gradient to 85% A). Combined fractions were concentrated to remove volatile solvents under reduced pressure, and the resulting solution was frozen and lyophilized to give the title compound (93 mg, 0.098 mmol, 28% yield) as a white solid. Major acetal isomer: LCMS (Method r, Table 7) Rt 0.83 min; MS m/z 945.4 . lH NMR (400 MHz, DMSO-d6) 8 9.78 (s, lH), 8.39 (d, J 7.2 Hz, lH), 8.13 (d, J 7.2 Hz, lH), 7.49 7.37 (m, 4H), 7.33 (t, J 2.2 Hz, lH), 7.28 (d, J 10.1 Hz, lH), 7.14 (t, J 8.1 Hz, lH), 7.10 7.05 (m, lH), 7.03 (s, 2H), 6.64 (dd, J 8.0, 2.4 Hz, lH), 6.13 (dd, J 10.1, 1.9 Hz, lH), 5.89 (d, J 1.5 Hz, lH), 5.50 (s, lH), 5.04 (s, 2H), 4.96 4.85 (m, 2H), 4.81 (s, lH), 4.55 (dd, J 18.1, 8.2 Hz, lH), 4.38 4.21 (m, 3H), 4.13 3.98 (m, 2H), 2.53 (dd, J 13.2, 5.2 Hz, lH), 2.28 (d, J 16.1 Hz, lH), 2.09 (d, J 11.2 Hz, lH), 2.08 1.95 (m, lH), 1.70 (dddd, J 29.9, 25.9, 14.4, 6.4 Hz, 5H), 1.36 (s, 3H), 1.26 (d, J 7.0 Hz, 3H), 1.18 (d, J 7.1 Hz, 3H), 1.02 (ddd, J 14.7, 11.6, 4.0 Hz, 2H), 0.86 (s, 3H).
Example 70: sis of2-((6aR,6bS,7S,8aS,8bS,l0R, l laR,12aS,12bS)(4-(3-((S)((S)(2-(2,5- Dioxo-2,5-dihydro-lH-pyrrol-l-yl)acetamido)propanamido)propanamido)benzyl)pheny1)hydroxy- 6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la, ,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2- d] [ l,3]dioxol-8b-yl)oxoethyl 2-(dimethylamino)acetate 2,2,2-trifluoroacetate Step 1: Synthesis of 2-((6aR,6bS,7S,8aS,8bS,l0R,llaR,12aS,12bS)(4-(3-((S)((S) ( (tert-Butoxycarbonyl)amino)propanamido)propanamido)benzyl)pheny1)hydroxy-6a,8a-dimethyl oxo-2,4,6a, 6b,7,8,8a,8b, l 12a,12b-dodecahydro-1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol-8byl )oxoethyl 2-(dimethylamino)acetate 2,2,2-trifluoroacetate 0 H o•·· Me2NCH2C02H HATU, 2,6-lut, DMF L .0 o r-""11 -- o 0 ''' H � BocHN/TI l�J�, 8 � 0 N HO BocHN� 0� � OH 0 = - O� F0o -N F F To a solution of tert-butyl ((2S)- l-(((2S)- l-((3-(4-((6aR,7S,8aS,8bS,l0R, l laR,12aS,12bS) hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b, l la, 12, 12a,12b-dodecahydro- 1H-naphtho[2', l':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)amino) oxopropanyl)carbamate (Prepared in same manner as Example 10, Step 1)(78 mg, 0.096 mmol), 2- (dimethylamino)acetic acid (10.9 mg, 0.106 mmol), and 2,6-dimethylpyridine (0.022 mL, 0.192 mmol) in ous N,N-dimethylformamide (2.0 mL) was added HATU (43.8 mg, 0.115 mmol), and the resulting solution was stirred at room temperature for 45 minutes. The crude product was purified by C, eluting with a solvent gradient of 5-95% MeCN in 0.lM aqueous TFA. Fractions containing the pure product were concentrated by lyophilization to afford the title compound (82 mg, 89% yield). LCMS (Method r, Table 7) R1 0.80 min, MS m/z +]. 898.2 [M+H Step 2: sis of R,6bS,7S,8aS,8bS,1ORl laR,12aS,12bS)(4-(3-((S)((S) (2-(2,5-Dioxo-2,5-dihydro-lH-pyrrol-l-yl)acetamido)propanamido )propanamido)benzyl)phenyl) hydroxy-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la, ,12b-dodecahydro-1H- naphtho[2',l ': 4,5]indeno[ 1,2-d] [ l ,3]dioxol-8b-yl)oxoethyl 2-(dimethyl amino)acetate 2,2,2- trifluoroacetate 0 0 .L_ --· 1) TFA/DCM o � (�( o 0 H 2) 0 N O -..,/"-�� �U-- 0»"� � r;, Boc N� 0N� 0 H ��o� J H 0 � H 0 0 0 0 O / F , t F, 0� -o i-Pr2NEt, DMF O F:x Fx - -N F ' - , N A solution of 2-((6aR7S,8aS,8bS,10R,1laR12aS,12bS)-l 0-(4-(3-((S)((S)((tertbutoxycarbonyl )amino)propanamido )propanamido)benzyl)pheny1)hydroxy-6a,8a-dimethyloxo- 2, 4,6a,6b, 7,8,8a,8b, l la, 12,12a,12b-dodecahydro-1H-naphtho[2', l ':4,5]indeno[l,2-d][l,3]dioxol-8b-yl) oxoethyl 2-(dimethylamino)acetate (82 mg, 0.074 mmol) in DCM (4 mL) and TFA (1 mL) was stirred at room temperature for 20 s, and then concentrated in vacuo. To a solution of this compound in anhydrous N,N-dimethylformamide (1 mL) was added Hunig's base (0.20 mL, 1.15 mmol) and 2,5- dioxopyrrolidin-l-yl 2-(2,5-dioxo-2,5-dihydro- lH-pyrrol- l-yl)acetate (27.8mg, 0.11 mmol). The resulting mixture was stirred at room temperature for 15 minutes, and TFA (0.106 mL, 1.376 mmol) was added. The crude product was purified by C, eluting with a solvent gradient of 5-95% MeCN in 0.lM aqueous TFA. Fractions containing the pure product were trated by lyophilization to afford the title compound as a colorless solid (46 mg, 0.0439 mmol, 59% yield). LCMS (Method r, Table 7) major acetal isomer R1 0.82 min, MS m/z +]; minor acetal isomer R 934 [M+H 1 0.81 min, MS m/z 934 [M+H+], 1H NMR (501 MHz, DMSO-d 6) 8 10.12 (s, 2H), 9.75 (s, lH), 8.40 (d, J 7.3 Hz, lH), 8.11 (d, J 7.1 Hz, lH), 7.45 - 7.42 (m, lH), 7.38 (dd, J 8.2, 2.0 Hz, 2H), 7.31 (d, J IO.I Hz, lH), 7.22 (d, J 8.2 Hz, 2H), 7.17 (t, J 7.8 Hz, lH), 7.06 (s, lH), 6.89 (d, J 7.7 Hz, lH), 6.50 (s, lH), 6.15 (dd, J .1, 1.9 Hz, lH), 5.93 - 5.90 (m, lH), 5.52 (s, lH), 5.30 (d, J 17.7 Hz, lH), 5.00 (d, J 17.7 Hz, lH), 4.86 (t, J 5.0 Hz, 2H), 4.36 - 4.25 (m, 4H), 4.12 -4.02 (m, 2H), 3.87 (s, lH), 2.82 (s, 3H), 2.56 - 2.51 (m, lH), 2.50 (s, OH), 2.50 (d, J 1.8 Hz, OH), 2.33 - 2.26 (m, 2H), 2.15 -2.06 (m, 2H), 2.04 - 1.97 (m, 2H), 1.84 -1.80 (m, lH), 1.77 - 1.60 (m, 4H), 1.37 (s, 3H), 1.26 (d, J 7.1 Hz, 3H), 1.19 (d, J 7.1 Hz, 3H), 1. 10 - 0.98 (m, 3H), 0.89 (s, 3H).
Example 71: Synthesis of 4-(2-( (6aR,6bS,7S,8aS,8bS,1ORl laR,12aS,12bS)- l 0-(4-(3-((S)((S)(2-(2,5-Dioxo-2,5- dihydro-lH-pyrrol- l-yl)acetamido )propanamido namido )benzyl)phenyl)hydroxy-6a,8a-dimethyl- 4-oxo-2,4,6a,6b,7,8,8a,8b,l la,12,12a,12b-dodecahydro-1H-naphtho[2',l':4,5]indeno[l,2-d][l,3]dioxol- 8b-yl)oxoethoxy)oxobutanoic acid Q\\\'' ytJ)y�)LND»''''. O 0 H H 0 o = Step 1: sis of 2-((6aR,6bS,7S,8aS,8bS,1OR,llaR,12aS,12bS)(4-(3-((S)((S) ( (tert-butoxycarbonyl)amino)propanamido)propanamido)benzyl)phenyl)hydroxy-6a,8a-dimethyl oxo-2, 4,6a,6b,7,8,8a,8b,l 12a,12b-dodecahydro-1H-naphtho[2',l':4,5]indeno[l,2-d][l,3]dioxol-8byl )oxoethyl tert-butyl succinate H02CCH2CH2C02tBu ·'-- ' HATU, t. ,-· H � (''>( l NJ� BocHN.,...If' , � The title compound was prepared using the method described for Example 70, substituting 4- (tert-butoxy)oxobutanoic acid for 2-(dimethylamino)acetic acid. LCMS (Method r, Table 7) R1 1.03 min; MS m/z +]. 968 [M+H Step 2: Synthesis of 4-(2-((6aR,6bS,7S,8aS,8bS,1OR,llaR,12aS,12bS)- l0-(4-(3-((S)((S)- 2-(2-(2,5-Dioxo-2,5-dihydro- lH-pyrrol-l-yl)acetamido)propanamido)propanamido)benzyl)phenyl) hydroxy-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la,12,12a,12b-dodecahydro-1H- naphtho[2', l':4,5]indeno[1,2-d][l,3]dioxol-8b-yl)oxoethoxy)oxobutanoic acid '--01•· 0-•· , 1) TFA/DCM H O � l NJ� 0 2) 0 0 BocHN.,... !( � 0 -\ �;lJo� 0 0 iPr2NEt, DMF The title compound was ed using the method described in Step 2, Example 69. It was isolated as a colorless solid (49 mg, 43%). LCMS (Method r, Table 7) R1 0.88 min; MS m/z 948.9 [M+H+]. 1HNMR (400 MHz, DMSO-d 6) 812.23 (s, lH), 9.74 (s, lH), 8.37 (d,J 7.3 Hz, lH), 8.09 (d,J 7.2 Hz, lH), 7.42 7.33 (m, 3H), 7.31 7.25 (m, lH), 7.17 (dd, J= 20.7, 7.9 Hz, 3H), 6.90 6.84 (m, lH), 6.15 6.09 (m, lH), 5.90 5.87 (m, lH), 5.48 (s, lH), 5.07 (d, J 17.7 Hz, lH), 4.86 4.79 (m, 2H), 4.37 4.23 (m, 3H), 4.12 3.98 (m, 2H), 3.85 (s, 2H), 2.65 2.58 (m, 2H), 2.52 2.47 (m, 2H), 2.32 2.24 (m, 2H), 2.09 (d, J 10.8 Hz, 2H), 2.02 1.94 (m, 2H), 1.85 1.56 (m, 6H), 1.36 (s, 3H), 1.24 (d, J 7.1 Hz, 3H), 1.17 (d, J 7.1 Hz, 3H), 1.10 0.95 (m, 3H), 0.85 (s, 3H). e 72: Synthesis of 2-((6aR6bS,7S,8aS,8bS,1 OR, llaR,12aS,12bS)- l0-(4-(3-((S)((S)(2-(2,5-Dioxo-2,5- dihydro-lH-pyrrol- l-yl)acetamido )propanamido )propanamido )benzyl)phenyl)hydroxy-6a,8a-dimethyl- 4-oxo-2,4,6a,6b,7,8,8a,8b,l la,12,12a,12b-dodecahydro-1H-naphtho[2',l':4,5]indeno[l,2-d][l,3]dioxol- 8b-yl)oxoethyl hydrogen sulfate Step 1: Synthesis of R,6bS,7S,8aS,8bS,10RllaR12aS,12bS)(4-(3-((S)((S) ( (tert-Butoxycarbonyl)amino)propanamido )propanamido )benzyl)pheny1)hydroxy-6a,8a-dimethyl oxo-2,4,6a,6b,7,8,8a,8b, l la,12,12a,12b-dodecahydro-1H-naphtho[2',l':4,5]indeno[l,2-d][l,3]dioxol-8byl )oxoethyl hydrogen sulfate To a solution of tert-butyl ((S)- l-(((S)- l-((3-(4-((6aR6bS,7S,8aS,8bS,lOR,l laR,12aS,12bS)- 7-hydroxy-8b-(2-hydroxyacetyl)-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l 12a,12b-dodecahydro- 1H-naphtho[ 2', l':4,5]indeno[l,2-d][l,3]dioxol- l0-yl)benzyl)phenyl)amino)oxopropanyl)amino) panyl)carbamate, prepared in a similar manner to Example 10, Step 1,(53 mg, 0.065 mmol) in MeCN (2 mL) was added pyridine sulfur trioxide x (42 mg, 0.26 mmol). The re was stirred at room temperature for 2 hours. The crude product was purified by C18 HPLC, eluting with a solvent gradient of 5-95% MeCN in O.lM aqueous TFA. Fractions containing the pure product were concentrated by lyophilization to afford the title compound. LCMS (Method r, Table 7) R1 0.83 min; MS m/z 892.0 [M+H+].
Step 2: sis of 2-((6aR,6bS,7S,8aS,8bS,1OR l laR12aS,12bS)(4-(3-((S)((S) (2-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l-yl)acetamido)propanamido)propanamido)benzyl)phenyl) hydroxy-6a,8a-dimethyloxo-2,4,6a,6b,7,8,8a,8b,l la,12,12a,12b-dodecahydro-1H- naphtho[2',l':4,5]indeno[l,2-d][l,3]dioxol-8b-yl)oxoethyl hydrogen sulfate �o �o r o rO O ,ro,� ') ;�:� HJ, I iyl )�V) ,io,,0 BocHN���-' ,,OJ) o --s'° 0 0 0 rN.
,,, H,, "1 = H 0=,7 OOH iPr2NEt, DMF , The title compound was prepared using the method bed in Step 2, Example 69. It was isolated as a colorless solid (27 mg, 28% yield). LCMS (Method r, Table 7) Rt=0.77 min; MS m/z = 928.9 [M+H+]. 1H NMR (400 MHz, DMSO-d 6) 8 9.75 (s, lH), 8.37 (d, J= 7.3 Hz, lH), 8.10 (d, J= 7.2 Hz, lH), 7.44 (d, J= 8.3 Hz, lH), 7.40 7.32 (m, 2H), 7.33 7.29 (m, lH), 7.27 (d, J= 10.1 Hz, lH), 7.23 7.12 (m, 3H), 7.04 (s, lH), 6.93 6.83 (m, 2H), 6.12 (dd, J= 10.1, 1.9 Hz, lH), 5.91 5.86 (m, lH), 5.42 (s, lH), 4.87 (d, J= 5.1 Hz, lH), 4.84 (s, lH), 4.74 (d, J= 18.3 Hz, lH), 4.45 (d, J= 18.3 Hz, lH), 4.36 4.24 (m, 3H), 4.11 3.99 (m, 2H), 3.86 (s, 2H), 2.58 2.48 (m, lH), 2.32 2.22 (m, lH), 2.08 (d, J= 11.1 Hz, lH), 1.98 (s, lH), 1.77 (s, 2H), 1.75 1.56 (m, 4H), 1.36 (s, 3H), 1.24 (d, J= 7.1 Hz, 3H), 1.17 (d, J= 7.0 Hz, 3H), 1.10 0.95 (m, 2H), 0.83 (s, 3H).
Example 73: Conjugation Protocols General Cysteine Conjugation Protocol An approximate 10 mg/mL solution of the desired antibody was ed in PBS buffer, pH 7.4 as well as a 10 mM TCEP solution in PBS (Pierce Bond-Breaker, cat. 77720). Antibodies (anti-hTNF hlgGl (D2E7) or anti-mTNF mlgG2a (8Cl l; McRae BL et al. J Crohns Colitis 10 (1): 69-76 (2016)) were then partially reduced by adding approximately two molar eq of 10 mM TCEP, briefly mixing, and incubating for 60 min at 37 °C. DMSO was then added to the partially reduced antibodies in sufficient quantity to 15% total DMSO. For the conjugations, 8 molar eq of a 10 mM leimide solution (wherein SM is a radical of a glucocorticosteroid and Lis a linker) were then added and incubated for 30 min at room temperature. Excess combo and DMSO were then removed using NAP-5 desalting s (GE Healthcare, cat. 1702) previously equilibrated with PBS , pH 7.4. Desalted samples were then analyzed by size exclusion chromatography (SEC), hobic interaction chromatography (HIC), and reduced mass spectrometry. ccinimide ysis 0 0 0 0 A-S-I N�N L D H H S �N�N L D 0 OH Hydrolysis of the thiosuccinimide ring of ADCs of the disclosure was accomplished by ting the ADCs at an elevated pH. Briefly, a 0.7 M arginine, pH 9.0 solution was prepared and added to each ADC in PBS buffer to bring the total arginine concentration to 50 mM (pH -8.9). The material was then incubated at 25°C for 72 hours. Hydrolysis of the succinimide ring was then confirmed by reduced mass spectrometry, after which, ysis was quenched with the addition of a 0.1 M acetic acid solution to 12.5 mM total acetic acid (pH - 7.1).
General Lysine Conjugation Protocol An approximate 10 mg/mL solution of the desired antibody was lly prepared in PBS buffer, pH 7.4. Eight molar eq of the D-L-N-hydroxysuccinimide (wherein SM is a radical of a glucocorticosteroid and Lis a linker) was then added to the antibody and incubated at 23°C for up to 24 hours in the ce of 15% DMSO. Conjugated samples were then desalted to remove excess combo and DMSO using NAP-5 desalting columns (GE Healthcare, cat. 1702) equilibrated with PBS buffer, pH 7.4. Desalted samples were then analyzed by size ion chromatography (SEC), hydrophobic interaction chromatography (HIC), and reduced mass spectrometry.
ADC Analytical Procedures Hydrophobic Interaction Chromatography. ADCs were profiled by hydrophobic interaction chromatography (HIC) to determine degree of conjugation and to calculate approximate drug to antibody drug ratios (DARs). Briefly, 100 µg of the ADCs were loaded onto an te 3000 Dual LC system (Thermo Scientific) ed with a 4.6 X 35 mm butyl-NPR column (Tosoh Bioscience, cat. 14947).
ADCs were loaded onto the column equilibrated in 100% buffer A and eluted using a linear gradient from 100% buffer A to 100% buffer B over 12 min at 0.8 mL/min, where buffer A is 25 mM sodium ate, 1.5 M ammonium sulfate, pH 7.25 and buffer B is 25 mM sodium phosphate, 20% isopropanol, pH 7.25. The DAR was determined by taking the sum of each peak percent area multiplied by their corresponding drug load and dividing the weighted sum by 100.
Size Exclusion Chromatography. Size distributions of the ADCs were profiled by size exclusion chromatography (SEC) using an Ultimate 3000 Dual LC system o Scientific) equipped with a 7.8 X 300 mm l 3000SWXL column (Tosoh Bioscience, cat. 08541). 20 ug of each of the ADCs were loaded onto the column and eluted over 17 min using an isocratic gradient at lmL/min of100 mM sodium sulfate, 100 mM sodium phosphate, pH 6.8 at 0.8 mL/min.
Example 74: ation ofadalimumab conjugated with a glucocorticosteroid to give an ADC Adalimumab MP-ala-ala steroid ADC having an average DAR 3 .5 was prepared by a twostep chemical process: disulfide reduction of adalimumab ed by alkylation (conjugation) with maleimidopropyl alanine-alanine steroid Cpd. No. 88.
OxNv-o�� I - ,, .. < o,,. '° '-o�� H ,H HN ·,,,, 0 �o Hr0{�o NH OH GN�O Cpd. No. 88 1] In the first step, a limited number of interchain disulfide bonds of adalimumab are reduced with tris(2-carboxyethyl) phosphine ("TCEP") (2: 1.8 equiv). Partially-reduced adalimumab is then conjugated to Cpd. No. 88 (2: 5 equiv) in DMSO.
Referring to Figure 5 which shows a chromatographic tion of the resultant ADC preparation, the ADC is a heterogenous mixture containing antibodies having zero drug linker molecules attached ("E0" peak), two drug linker molecules attached ("E2" peak), four drug linker molecules attached ("E4" peak), six drug linker molecules attached ("E6" peak) and eight drug linker molecules attached ("E8" peak), depending upon the number of interchain disulfide bonds reduced. s of tographically separating and isolating the homogenous E2 and E4 peaks are described by Hamblett et al., Clin Cancer Res 2004;10:7063-7070. The HIC conditions used in Figure 5 were as follows: The column was TOSOH Tskgel Butyl-NPR 4.6 mm x 3.5 cm, 2.5µ and the column temperature was 30°C. Wavelength was 280 nm, run time was 22 minutes, injection, volume was 40 µL, flow rate was 0.5 mL/minute. Mobile Phase A: 25mM Na2HPO4, pH 7.0 and 1.5M SO4, Mobile Phase B: 25mM Na2HPO4, pH 7.0/IPA =75/25. Gradient Profile: Time (minutes) Mobile Phase A Mobile Phase B 0 90 10 2 85 15 18 5 95 18.1 90 10 22 90 10 Methods of chromatographically separating and isolating the nous E2 and E4 peaks are described by Hamblett et al., Clin Cancer Res 2004;10:7063-7070. Briefly, after hydrolysis and adjustment to pH <7.4, the broad distribution mixture was treated with 3 M ammonium e/50 mM phosphate buffer to bring the overall solution tration ofammonium sulfate to approximately 0.8 M.
A pre-packed hobic Interaction Chromatography (HIC) column (resin butyl sepharose HP) was prepared by sanitizing with 0.5 N NaOH solution (4 CV), rinsing with water for injection (WFI, 0.5 CV) and equilibration with 0.8 M ammonium sulfate/25 mM phosphate buffer (4 CV). The broad distribution/ammonium e buffered on was loaded on the HIC column (approximate loading, 30 mg protein per mL of resin) followed by a wash with 0.8 M ammonium sulfate/25 mM phosphate buffer (2.5 CV). Elution of the product was as follows: 0.72 M ammonium sulfate/25 mM phosphate buffer (3 CV), unconjugated mAb; 0.56 M ammonium sulfate/25 mM phosphate buffer (4.5 CV), DAR2 ADC; 0.32 M ammonium sulfate/25 mM phosphate buffer (6.5 CV), DAR4 ADC. The DAR 2 and DAR4 product fractions were then separately concentrated to approximately 30 mg/mL via ultrafiltration pore Ultracel, 30 kD cutoff) followed by tration into WFI (8 CV).
The succinimide of the purified E4 conjugate was hydrolyzed to provide the stabilized attachment by ing the pH of the product solution to � 9 using an arginine buffer. The solution was held at ambient temperature for� 2 days at which time LC-MS analysis determined the hydrolysis was >90% te. See Figure 6 for a portion of the LC-MS chromatogram. The SEC conditions used in Figure 6 were as follows: The column was TOSOH TSK-gel G3000SWxL , 5µ, 250A, 7.8 x 300 mm, the column was ambient temperature, Wavelength was 214 nm, Run Time was 55 minutes, Injection Volume wasl0 µL, Flow Rate was 0.25 mL/minute, mpler Temp. . Mobile Phase: 100 mM Na2HPO4 & l00mM Na2SO4, pH 6.8 /IPA= 90/10.
Raw (Figure 7) and deconvoluted (Figure 8) MS data of umab conjugated with MP­ ala-ala- steroid Cpd. No. 88. Black square and circle represent the ADC with succinimide hydrolyzed and unhydrolyzed, respectively. The relative abundance of hydrolyzed and unhydrolyzed ADC is used to determine hydrolysis conversion.
Hydrolysis Hydrolysis of succinimide ring after conjugation was conducted with borate buffer at pH 8.0, pH 8.5 and pH 9.0 and arginine buffer at pH 8.0 and pH 9.0 to study the rate of ring ysis. The results are shown in Table 9 below. ???? ???? ???? ???? ???? ???? ???? ???? ?? ???? ???? ???? ???? ???? ???? ???? ???? ???? ???? ???? ???? ???? ???? ???? ?? ?? ? : ?????????? ?: ???????: ?: ???????: ???????: ???????: ???????: ???????: ???????: ? ? ? : ???0?????: ?? ?? ? : ? 4 ? : ? ? ?????????? ???????????? ?????????: ?????????: ???: ?????????: ?????????: ?????????: ? 1 ? 9 ? # ? 4 ? ’ ? : ? % ? + ? 7 ? ? ? 4 ?? ?? ? ) ? 9 ???????????? ???????????? ?????????: ???????: ???????: ???????: ?????: ?????????: ?????????: ???????: ???????: ?????????: ? - ?????????????? ???????????????? ? " ? / ? 2 ?? ?? ? % ?????????????? ???????????? ?????????: ?????????: ?????: ?????????: ?????????: ???????:???????: ?????????: ? 9 ? ? ?????: ?????????: ?? ???????: ?: ?? ???: ?????????: ?????????: ?????????: ?????????: ???????: ???????: ?????????: ???????????????????????? : ???????????? ? ? ?????????? ?????????????? ?????? ?????????: ?????????: ?? ???????: ?? ???????: ?????????: ?????????: ?????????: ???????: ???????: ???: ?????????: ? ? 0 ? ? : ? : ? ? ? ? ? ? ? ? 4 ?????????: ?????????: ?????????: ?????????: ?????????: ?????????: ?? ???????: ?? ? 4 ? 9 ? : ? : ?????????? ???????????? ?? ?????????? ?????????: ? + ? 1 ? ’ ? : ? % ? # ? 6 ? 4 ? ? ?????????: ?????????: ???: ???: ?????????: ???????????? ?????????: ?? ???????: ?? ? ) ? 9 ?????????????? ???????????? ?? ?? ? - ?????? ? / ? " ? 2 ?????????: ?????????: ?????????: ?????????: ?????????: ???: ???????: ???????: ???????????? ?????????: ?? ?? ? % ?????????????? ? 9 : ? ? ?????????: ?????????: ?????????: ?????????: ???????????? ?????????: ?? ???????: ???????: ?? ?????????: ?: ?????????: ? ? ?????????????? ? ? 0 ? ?????????: ?????????: ?????: ???????????? ?????????: ?? ???????: ?? ?????????: ???????: ???: ?? ? ?????????? ? : ? ? : ? ? ? ? ? ? : ?????????? ? : ???????????? ?????????: ?? ???????: ?? ?????????: ?????????: ?????????: ???: ?????: ?????????: ?????????: ?????????: ? 2 ? 8 ? 4 ? & ? " ? : ? $ ? * ? 5 ? ? ? 3 ?????????????? ?????????????? ???????????? ???????????? ?????????: ?? ???????: ?? ?????????: ?????????: ?????????: ?????????: ?????????: ?????????: ? 8 ?? ?????????: ?????????: ?????????: ???: ? ( ? ! ? , ? . ? 1 ?? ???????: ???????: ?????????: ?????????: ?????????: ? 8 ???????: ???????: ???????: ?? ???????? ???????: ???: ???????? ?????????: ?????????: ???????: ?????????: ???????? ? ? ???0?%???: ?????????????? ???????????? ?????????: ? ?.???: ???????: ?????: ?? ???????? ???????: ?????? ?????????: ???????? ?????????: Example 75: In vitro activity of small molecule steroids Glucocorticoid Receptor Binding Assay Small molecules were tested for glucocorticoid receptor (GR) binding using the Polarscreen™ Glucocorticoid Receptor Assay Kit, Red oFisher A 15898) according to the manufacturer's protocol. Briefly, compounds were serially diluted in DMSO then transferred into assay kit buffer at a 1: 10 dilution. Compounds were further diluted 1:5 in assay kit buffer, and 10 µI was transferred to a 384 well low volume black walled plate (Coming 4514). 5 µI of 4X Fluormone GS Red stock solution and Sul of 4x GR full length stock solution were added to each well containing test compound, and plates were ted protected from light at room temperature for 4 hours. Fluorescence Polarization (mP) was measured for each plate using an EnVision Multilabel Plate Reader (Perkinelmer # 2104-0010), and data were analyzed using a four parameter curve fit to te EC50 values. The results are shown in Table below.
Mineralcorticoid Receptor Cell Assay Small molecules were tested for lcorticoid receptor (MR) agonist ty using the PathHunter® NHRPRO CHO-Kl MR cell line (DiscoveRx cat# 93-0451C2) according to the manufacturer's protocol. Briefly, 20,000 cells/well in culture medium were plated in a 96 ell plate (Costar cat# 3885) overnight at 37°C. Media was removed and replaced with serially diluted small molecules in assay medium (30 µI; 0.3% DMSO final). Plates were ted overnight at 37°C. Media was removed, replaced with detection t (DiscoveRx cat# 93-0001; 12 µI/well), and incubated at room temperature (RT) for 60 minutes. Luminescence was measured for each plate using an EnVision Multilabel Plate Reader (Perkinelmer # 2104-0010), and data were analyzed using a four parameter curve fit to generate EC50 values. The results are shown in Table 10 below.
Progesterone Receptor g Assay 2] Small les were tested for progersterone receptor (PR) binding using a modification of the LanthaScreen® TR-FRET Progesterone Receptor Coactivator Assay (Thermofisher cat# Al5903) where the fluorescein-labeled coactivator peptide was replaced with Fluormone AL-Red (Thermofisher cat# PV4294) to improve assay . Briefly, compounds were serially diluted in DMSO, then transferred into assay buffer (Thermofisher cat# PV 4301 + 5mM DTT) at a 1: 10 on. 10 µI of compound was transferred to a 96 half-area black well plate (Coming cat#3694) in duplicate. 5 µI of PRLBD protein (4nM stock in assay buffer; Thermofisher cat# P2899) was added to each well. In addition 5 µI of a ed mixture of Fluormone AL-Red (12nM) and terbium-labeled anti-GST monoclonal antibody (mAb) (20nM; Thermofisher cat#PV3550) in assay buffer was also added to each well. Plates were incubated at room temperature (RT) for 2 hours, and then TR-FRET emission ratio was measured using an EnVision Multilabel Plate Reader nelmer # 010). Data were analyzed using a four parameter curve fit to generate EC50 values. The results are shown in Table 10 below.
Androgen Receptor Binding Assay Small molecules were tested for androgen receptor (AR) binding using a modification of the LanthaScreen® TR-FRET Androgen Receptor Coactivator Assay (Thermofisher cat# Al5878) where the fluorescein-labeled coactivator peptide was replaced with Fluormone AL-Red (Thermofisher cat# PV4294) to improve assay . Briefly, compounds were serially diluted in DMSO then transferred into assay buffer (Thermofisher cat# PV4295 + 5mM DTT) at a 1: 10 dilution. 10 µl of compound was transferred to a 96 half-area black well plate (Coming cat#3694) in duplicate. 5 µl of AR-LBD protein (5nM stock in assay buffer; Thermofisher 09) was added to each well. In addition 5 µl of a prepared stock of Fluormone AL-Red (20nM) and terbium-labeled anti-GST monoclonal dy (mAb) (30nM; Thermofisher cat#PV3550) in assay buffer was also added to each well. Plates were incubated at room ature (RT) for 6 hours then TR-FRET emission ratio was measured using an EnVision Multilabel Plate Reader (Perkinelmer # 2104-0010). Data were analyzed using a four parameter curve fit to generate EC50 values. The results are shown in Table 10 below.
GRE Reporter Assay K562 parental GRE (pGL4.36[luc2P/MMTV/Hygro]) cells described in Example 78 were plated onto 96 well tissue culture treated white plates (Costar: 3917) at 50,000 cells per well in 50 µL of assay medium (RPMI, 1 % CSFBS, 1 % amine, 1 % Na Pyruvate and 1 % MEAA). Small molecule GR agonist compounds were serial diluted at a starting concentration of 100 µM and serial diluted 4 fold in 100% DMSO. The small molecule compounds were diluted further in assay medium by transferring 2 µ1 of serial d compounds into 248 µ1 assay medium into a secondary dilution plate (1:125 dilution).
The cells were then treated with 25 µL of 1:125 diluted GR agonist compound for a final starting concentration of 266.7 nM (1:3) or media alone and incubated for 24 hours at 37°, 5% CO2 . After 24 hours tion, cells were treated with 75 µL of Dual-Glo Luciferase Assay System (Promega-E2920) for 10 minutes and analyzed for luminescence using the TopCount or \hcroH�.:Lt2 (PerkinElmer).
Estrogen Receptor Binding Assay Small molecules were tested for estrogen receptor (ER) alpha binding using a modification of the Screen® TR-FRET Estrogen Receptor Alpha Coactivator Assay (Thermofisher cat# Al5885) where the fluorescein-labeled coactivator peptide was replaced with Fluormone ES2 Green (Thermofisher cat# PV6045) to improve assay signal. y, nds were serially diluted in DMSO then transferred into assay buffer (Thermofisher cat# PV4295 + 5mM DTT) at a 1: 10 dilution. 10 µl of compound was erred to a 96 half-area black well plate (Coming cat#3694) in duplicate. 5 µl ofERLBD protein (5nM stock in assay buffer; Thermofisher cat#4542) was added to each well. In addition 5 µl of a prepared stock ofFluormone ES2 Green (12nM) and terbium-labeled ST monoclonal antibody (mAb) (8nM; Thermofisher cat#PV3550) in assay buffer was also added to each well. Plates were incubated at room temperature (RT) for 4 hours, and then TR-FRET emission ratio was measured using an EnVision Multilabel Plate Reader (Perkinelmer # 2104-0010). Data were analyzed using a four parameter curve fit to generate EC50 . The results are shown in Table 10 below.
'"Cl � � -- � g � Ut Ut '""' QC) :;:; 8 � -- � Z: ;:J NT NT AR Binding ICso (µM) NT >30 I 3.99 I NT NT ER g ICso (µM) NT I 30++ >30 I NT NT PR Binding ICso (µM) NT 0.0026 I 0.0198 I NT NT ) MR ( Agonist ECso (µM) NT 0.0846 08 36 0 : 957 I I 0.0184 0.0396 GRE Rep orter ECso (µM) NT 0.0002 0.0003 0.0120 0.0641 I I - GR IC� o - 312 in vitro activity I binding 0.0066 0.0036 I 0.0095 I Table 10: Q r0i"l� 0 2 � NH 0H h �NH 2 - s � �N H2 I: ' t rue ure OH �s 1 ---� r(Y"S . 1 St em1ca 0 � , �O H�_ 0� __ OH 0 TFA OH o � ,o �I TTA r('''��t·lo' .. ,o o ... •0 i ·" o.,__ ) .... ) . . o .:/_.,,v Ch A "''� � � 0 �� 0 F � l 1 l C F _ ':" H2N � 0 o -- 'v' � 0 I I ) I I . I A ) ) I ( T ; 3 ( TFA 4 5 Cpd No . 6 ( TFA :;; 0 N 0 -- 0 (,,; Ut Ut """ QC) """ ---l -- N """ 0 ""' ---l """ -- 00 N 0 """ ---l "'d ("") I AR g I ICso (µM) >30 NT I >30 >30 NT I I ER Binding I ICso (µM) >30 NT I 30++ 5.13 NT I I PR Binding ICso (µM) 0.0157 NT 0.0116 0.0105 NT ) I M R ( Agonist ECso I (µM) 0.515 0.300 NT I 2.61 0.305 NT I I GRE Reporter ECso (µM) 0.0005 0.0151 I 0.0001 I 0.0001 0.0185 I I -313 - GR binding IC�o 0.0155 0.0201 0.0094 0.0156 0.0139 I I I I I o-NH2 - ,a, ,a, s n b H 2 N p s n A --<' a 0 us o-NH2 --<' OH 7 ,,,Q-s � .. ,, OH ) ... ,0 >··• .,.,0 OH ) OH )p OH 0 o� l F o�) _;y-s Chemical Structure o � �:�;?"� o � . o�) - lj : F �:�0 : F l : i= _ I --...:;,- ":' _, 1 � l ) ..,o "Y i = 0 ,etlfP: ..;? - 0 o·.,- 0 o.,,,.. 'v'" I I I I I I 9 10 11 7 Cpd. No. :;; 0 N """ 0 ""' ---l """ "'d -- ('.j 00 N 0 """ ---l -- 0 (,,; Ut Ut """ QC) N 0 """ ---l -- I AR Binding ( M) NT >30 >30 >30 NT IC so I I I I ER NT NT I >30 2.71 3.77 I I I PR NT 0.0154 0.0222 0.0165 NT R ) M ( M) NT 10.1 >30 0.119 0.141 NT EC so I I I I ( Agonist GRE Reporter ( M) 0.0161 0.0009 0.0001 0.0006 0.0188 EC so I I I I I - GR binding 0.0247 �o o . 0157 0.0255 0.0149 0.0537 IC - 314 I I I I I I 0 u:s I C l N H2 :::,... � I N H 2 Cl I N H 2 Cl s H2 N 9' o-: s '7 � s '7 9' � � Cl N H 2 A A •" OH .. ,, 0 7 s ••0 " A ., .. 0 7 · '7 "' A 9' ""0 7 · O H o�) o�) .... 0 7 · l >- ••O "O 1 >·• • O o�) {X: 1 >• •L O o�) -M 1 > · • • O i = OH 0� 1 ':1 - I i = I j ':1 i= OH 1 ':1 -y i = O H al Structure - I o - .,. � - I j ':1 j >- i = - I j ':1 -...:;,, y -...:;,, y o ·.-, o ·.., I I I I I Cpd. I No. 12 13 14 15 16 :;; 0 -- Ut """ QC) N 0 """ ---l -- N """ 0 ""' ---l """ 0 Ut "'d ("") -- 00 N 0 """ ---l (,,; I AR g IC so (µM) NT >30 >30 NT NT I I I ER Binding IC so (µM) NT 30++ 0.814 NT NT I I I PR Binding IC so (µM) NT 0.0171 0.0186 NT NT ) MR 1.64 so ( Agonist EC (µM) NT 1.64 0.521 NT NT I I I GRE Reporter EC so (µM) 0.0152 0.0002 0.0003 0.0656 0.0299 I I I - GR IC�o - 315 binding I 0.0101 0.0194 0.0086 I I 0.0278 0.0437 I I NH2 � - NH2 NH2 NH 2 NH2 A◊ f I ,,:::: I ,,:::: :::,._ Cl :::,._ Cl 7' I 7' I TFA & � Chemical Structure TFA � (' :::,... o � _,, :::,... - l OH ,.Q OH o°') -hs >·•• O l ...o >-•• O l n, j .. ,o P -- 0 .,,0 OH . HO T\.,, HO. 1 1:1 o°') _p-;s 1 1:1 =. F F F - I 0� i= y - I 0� i= &P'o rO--q .1 H O --....:;,,, -y � Or' � o- ,, Cpd. I I I I I No. 17 18 ) ) I 1 9 ( TFA 2 0 2 0 ( TFA :;; 0 N 0 '""' � '""' 0 ""' ---l '""' "'d (""l g 00 N 0 '""' g (.,; Ut Ut '""' QC) AR Binding M) IC so ( µ >30 NT NT NT >30 ER Binding M) IC so ( µ >30 NT NT NT 30++ PR Binding M ) IC so ( µ 0.0073 NT NT NT 0.0089 R ) M M) ( t EC so ( µ 0.149 4.42 NT NT NT 1.06 GRE Reporter M ) EC so ( µ 0.0002 0.0075 0.0233 0.0051 0.0023 - GR binding M ) IC so ( µ 0.0283 0.0082 0.0094 0.0108 - 316 0.0138 � - ?:> 1 ,,::;, NH 2 f p � H2 N IA H2 N f - :::,.. :::,.. TFA Chemical Structure OH ....0 7 ""0-0 .,,o pv'� pv""; ,,,0 OH , o :rO- ·•• 0 OH ·• • 0 0 0 ' ""OT"°q 0 H . 0 � ,o � �I .. ,0 H H ' F ,o F ' ' , H , o F F . . F F , _ F 9' ✓,; ,o # HO # 0 � �1 9' 0 O 9' 0 O 22 ) ) ( TFA 22 23 24 ( TFA Cpd. No. 21 :;; 0 g Ut """ QC) N 0 """ ---l N """ 0 ""' ---l """ (.,; Ut "'d ("") g 00 N 0 """ AR Binding IC so (µM) >30 NT NT NT ER Binding IC so (µM) 30++ NT NT NT I I PR g IC so (µM) 0.0133 NT NT NT ) MR ( Agonist EC so (µM) 3.09 NT NT NT GRE Reporter EC so (µM) 0.0002 0.0167 NV 0.0419 - GR IC � o - 317 binding 0.0216 I 0.0102 0.0146 I 0.0256 C(U NH 2 Cl .,.-:; I :::,,_ _..,. NH ll � _......N H ,, � ✓,; Chemical Structure ,, ,o OH ·· 7 . . Of ,0 7 OH ""0 ff-P'C � NH 2 �y OH >� O o,.. ) . - ftf ti : F F : F �H F 0 0 ~ I 0 I I I I 26 27 28 Cpd. No. 25 :;; 0 N """ 0 ""' ---l """ "'d (""l -- 00 N 0 """ ---l -- 0 (.,; Ut Ut """ QC) N 0 """ ---l -- AR Binding ( M) NT NT NT >30 IC so ER NT NT NT 30++ I I I PR NT NT NT 0.013 R M (Agonist) M) ( NT NT NT 3.56 EC so GRE Reporter ( M) 0.0003 0.0001 EC so I 2 0.0011 0.0078 - 0.0074 GR o IC � 0.013 0.0177 0.0107 - 318 9 binding : - 2 H NH I . N � 9 1/"' :::,... H 1/"' I H .., � I /4> 'N /4> 'N :::,... OH OH "0 7 . , , o OH ) . , , o " 0 OH �A F i= ' F F Chemical Structure 0 �A � 0 � 0 I I I I Cpd. I No. 29 30 31 32 AR Binding >30 so ( M) NT NT NT IC I I ER 30++ NT NT NT I I PR 0.256 NT NT NT R ) M 0.407 0.407+ M) so ( NT NT NT EC I I ( Agonist GRE 0.0178 er so Report ( M) 0.0306 0.0034 0.0214 EC I I I - GR ng IC �o - 319 bindi 0.0169 0.0122 0.0034 0.0067 NH 2 : , 9 _,....N H : � 2 NH � 'o � � � N TFA OH OH '° N o O A A OH V .. l - r ., ,o O H 1 ... · • 1 0 F F .., o 0 lj al Structure o � o � H0 � ,0 I 0 I I I ) I Cpd. I No. 33 34 35 35 ( TFA AR Binding IC so (µM) >30 >30 9.28 >30 >30 I I I I ER Binding IC so (µM) >30 >30 >30 30++ >30 I I I I PR Binding IC so (µM) 0.052 1.09 1.19 0.137 0.106 ) MR ( Agonist EC so (µM) 0.129 >30 2.12 5.05 1.47 0.422 >30 >30 I 0.876 I I GRE Reporter EC so (µM) 0.0002 0.1320 I 0.266++ 0.0322 I 0.0007 I - GR IC � o - 320 binding I 0.0034 0.0131 I 0.0128 0.0156 I 0.0111 I , N H ; 2 P N H � N H 2 N H 2 I� 4 N H � P � - � � A' du TFA � I N "° N "° .0 ' . - al Structure I V O (r I O H .,, o O H ., r OH •' .,,o- r ·'' � 0 .,,0 , ,, o O H .,,o- , o ; .. , a 0 0 �l ' i = H O � H o �J A 0 0 0 �,o 7·•'' a-;.,,�...._.... 0 I I I I I 38 37 39 ) I Cpd. No. 36 ( TFA 40 AR g IC so (µM) >30 >30 >14.42 >30 I I ER Binding IC so (µM) 30++ 9.75 6.48 30++ I I PR Binding IC so (µM) 0.076 0.0873 0.08 2.13 ) MR ( Agonist EC so (µM) 30++ 10.1 30++ >10 30++ I I GRE Reporter EC so (µM) 0.0014 0.0004 0.0013 0.0541 I I - GR binding o 0.0028 0.0124 o.0661 0.0267 IC � - 321 I I I ; ,0 NH NH 2 NH; I ,,;:; ·''::::,... VVNH2 I TFA oi? TFA . ' , Chemical Structure OH .,,0 ,,, :::,.,. 7 o � � __,,01 H JH o� � o--Q ,o-l • O - - fiBP•O v'QJ &7 '.,,0 i= F 0 &JP � 0 0 0 ) I I ) I I I 4 5 ( TFA 46 ( TFA No. 41 Cpd. 42 I >30 AR ICso (µM) NT NT NT I Binding I I I 30++ ER ICso (µM) I Binding I >30 I >30 I >30 0.0345 PR ICso (µM) 2.7 I Binding I 0.171 I I 0.108 10 1 ) 0 7 5 1 · M R ECso (µM) 3.86 >30 >30 I ( Agonist I I I 0.0009 GRE Reporter ECso (µM) 0.0080 I I I 0.0042 I 0.0183 I 0.0871 - GR IC�o . - 322 binding I I I 0.0065 I 0.0056 I I 0.0118 NH2 NH2 h(l NH 2 � I NH2 -9' rue ure t C(V CV-OH �S l(V HO ,,V . 1 St . • ' . . o . . o OH 7·· 0 ,,10 Ch em1ca HQ �_] HO. HO. O.:-..) ' i= 01··' =-�.--01·•' un un ......,,, . ·- Y ..f'-.,, 0 o � o � o·, -..:;,r 47 I I I I 48 49 Cpd. No. so I AR Binding (µM) >30 NT NT >30 IC so I I I ER Binding (µM) 30++ NT NT >30 IC so I I I PR Binding (µM) 0.0136 NT NT 0.0527 IC so I I I ) M R (µM) 4.98 NT NT 0.269 6.23 EC so I I I ( Agonist GRE 0.0003 Reporter (µM) 0.0009 0.0006 NT EC so I I I I GR g 0.0065 0.0095 NT IC �o I I 0.0042 I I I NH, 2 2 0 TFA HO () N H � ' ,,.._ ,, 7 . , ' .,,0 0 .. ,o .,,0 0 .,,0 HO. tiBP. . - . - ' HO. _p>-q HO F . F HO�'l i = Chemical Structure 0�p-q 0 � 0 �./ . 0 I I I ) I Cpd. I No. 51 ( TFA 52 53 54 I AR NT IC so NT NT >30 >30 I (µM) I Binding I I I ER so I >30 IC (µM) >30 5.98 >30 30++ I Binding I I I PR so I 0.0699 IC (µM) I Binding 0.0134 0.0341 0.0933 0.0914 ) I I I >30 MR EC so (µM) 0.179 10.5 0.830 >30 10.1 I I ( Agonist I I GRE 0.0017 so I Reporter EC (µM) 0.0004 0.0013 0.0004 0.0015 1 I I I - GR 1 IC �o - 324 binding I 0.01 2 I 0.01 2 I 0.0078 I 0.0114 0.0063 I H2 NH 2 A N H; NH 2 � H, N o I� Q ::::,.... A vO �OH SV I 0 S VN TFA I o V .,,v:s Chemical ure ,,, · OH '0 · ,,, ,,V 7 ·"' · ·' .,,o OH o.-....) 0 7 y·\' .,, o OH ....07 OH •IIQ OH 07 •IIQ H o .-.... ) ) . 0---- F "f\'07 : F ··-y - L.J ··-y - U y't'"\' . ·- - LI o � &•O 0 & o.....-�-.............. 0 I I I I I I 58 ) Cpd. No. 55 ( TFA 57 59 60 I I AR g IC so (µM) NT NT NT NT NT I I ER Binding IC so (µM) >30 >30 >30 >30 NT I I I I PR Binding IC so (µM) 0.839 0.0248 0.447 0.121 NT ) MR so ( Agonist EC (µM) >30 0.475 >30 30++ NT I I GRE Reporter EC so NT (µM) 0.0191 0.0030 0.0067 0.0214 I I - GR IC�o NT - 325 binding 0.0302 0.0210 I 0.0038 0.0116 I I I I SUNH2 Cl NH2 Cl NH 2 b I ,O' I b ---:: I ,,,-:::. b S O UNH 2 S V ONH, g o V d ¥ , Chemical Structure ,,,U ,,, ,,, ,,, OH •1 1 0 - OH 0 •IIQ 7· O H => 0 7' .,, o "Y°'\1 07· o Q 1•07· .,, OH O H 1 O ••I . - r'\ p :> . - 1 1 LI nu� · ·~y - _1 .......,,, 0 � q: 0 � q O '/ '-,1/ I I I I I I 63 64 65 62 61 Cpd. No.
I I AR Binding ICso (µM) NT NT NT NT I I I I ER Binding ICso (µM) NT 30++ NT 30++ I I I I PR Binding ICso (µM) NT 0.181 NT 0.0096 MR (Agonist) ECso (µM) NT 30++ NT 30++ I I I I GRE er ECso (µM) NT 0.00313 NT 0.00244 I I I I - GR IC� o NT 0.012 NT - 326 binding I I I I I 0.00749 NH 2 NH2 Structure "" 1 ds� -" NH, ((U-NH2 S � •' VO � HO ·' o H 7 .,,0 7' .,,0 F OH ..,,0 )- .. o . - ( s cr-o . - �) Chemical @3:�J" HO. 0 l ' F �l F 1 t1 _ 1 F 0 � 0 0 r /........:;,,,,"'::' o I I I I I 67 68 69 No. 66 AR Binding ( M) NT data ICso ER NT reported the PR NT than" R "greater ) M M) as ( NT read be ( Agonist ECso GRE Reporter M) can ( 2 data ECso I - GR reported �.. ·s yr, - 327 0.0198 and the �... points data S oNH2 of multiple OH ... ,o p ,,, o an average is i = data Chemical Structure 0 reported that the Not Tested Cpd. No. 70 ++ indicates NT indicates Example 76: Stability ofanti-TNF-alpha immunoconjugates Matrix Stability Anti-TNFa steroid ADCs were tested for their susceptibility to prematurely release small molecule d under physiological conditions. In these experiments, ADCs were diluted in plasma (human, monkey, mouse, or rat) or buffer in ate and ted for 6 days at 37°C, 5% CO2 . Each sample was quenched at time 0 minutes and at various time points over the 6-day period. Samples were then analyzed using LC/MS/MS and compared with rd curves for the ponding small molecule. The % maximum release of small molecule payload over time was calculated. The results are ized in Table 11 below.
Table 11: Stability ofanti-TNFa steroid ADCs Matrix ity (% max SM release) Cpd. Cynomolgous Monkey PBS buffer Human plasma Mouse plasma No. Plasma 136 0.00864 8.34E-04 0.0165 0.0327 137 0 0 0 0 138 0.00954 0.00471 0.00444 0.0215 139 0 0 0 0 142 0.00306 0 0.00766 0.0564 144 0 0 0 0 145 0.00451 0 0 0 146 0.031 0.016 0.0306 0.0744 147 0 0 0 0.00565 148 4.51E-04 0.0032 0.0188 0.0162 149 0 0 0 0.00648 150 0.00809 0 0.0153 0.074 152 0.00513 0 5.42E-04 0.0392 153 0 4.0lE-04 0 0.132 154 0.00E+00 0 0 0 155 0 0 0 0 156 0 0 0 0.152 157 0.0134 0.00559 0 0.026 These results demonstrate that anti-TNFa d ADCs are stable in buffer and plasma of multiple species and that minimal small molecule release is observed.
Proteolytic Stability The susceptibility of steroid ADCs to release their payload through protease treatment was compared with an ADC generated using the vcmcMMAE drug linker conjugated to a murine CD-19 antibody. ADCs (average DAR of 4) were incubated with either cathepsin B or nase K, and payload release was analyzed by LC-MS at various timepoints (0, 1, 4, 7 and 24 hours).
The results are shown in Figure 1 and demonstrate that the steroid ADCs are resistant to ous cathepsin-mediated release of payload from the ADC. This is in contrast to a known payload linker (mcvcMMAE) ADC, where MMAE is released in significant amounts upon cathepsin treatment.
This data indicates that steroid ADCs are much less susceptible to premature payload release that s from cathepsin activity in circulation than known ADCs. Indeed, steroid release is only observed with proteinase K, a serine protease that displays broad cleavage specificity. This indicates that the antibody portion of the steroid ADC needs to be significantly lized prior to d linker cleavage and that d release can be restricted to an environment where digestion of the antibody scaffold of the ADC can occur, such as the lysosome.
Cathepsin B Digestion A mL stock solution of cathepsin B (Sigma) was prepared in buffer (25mM Tris, 50mM NaCl and 5% glycerol). To generate a 10 µg/mL working on of cathepsin B, 5 µl of 0.2 mg/mL cathepsin B stock was mixed with 95 µl of activation buffer (50mM sodium acetate pH5, lmM EDTA, and 5mM DTT) and incubated at 37°C for 15 minutes. For ADC digestion, 20 µl of 100 ug/mL ADC and 20 µl of sin B working solution were mixed with 160 µl dilution buffer (50mM sodium acetate, lmM EDTA). The sample was incubated at 37°C with shaking, and 40 µl aliquots were removed after 0, 1, 4, 7, and 24 hours. To each aliquot was added 160 µl ofquench solution (0.1% formic acid; 1: 1 MeOH:MeCN; 100 nM carbutamide), and released small molecule was detected by LC-MS/MS as previously described.
Proteinase K Digestion A 5 mg/mL stock of proteinase K (Sigma) was prepared in deionized (DI) water. A 0.25 mg/mL working solution of proteinase K was prepared by mixing 50 µL of 5 mg/mL proteinase K with 950 µl dilution buffer (lxHBSS and lmM EDTA). For ADC digestion, 20 µL of 100 ug/mL ADC and 40 µl of nase K g solution were mixed with 140 µl dilution buffer. The sample was incubated at 37°C with shaking, and 40 µL aliquots were removed after 0, 1, 4, 7, and 24 hours. To each aliquot was added 160 µl of quench solution (0.1% formic acid; 1:1 MeOH:MeCN; 100 nM carbutamide), and released small molecule was detected by MS as usly described.
Example 77: In vivo stability of anti-TNF-alpha immunoconjugates The tibility of the steroid ADC to undergo drug linker loss was assessed in mice. MP-Ala- Ala-steroid was conjugated to human IgG 1 mAb (av. DAR 4) and incubated at pH 9 to catalyze ringopening ysis of the thiosuccinimide ring. After neutralization, the steroid ADC was injected in mice, and the kinetics of drug linker loss were monitored over 7 days by LC-MS.
In these ments, ADC formulated in phosphate buffer saline was dosed intravenously to 15 male DBA/1 mice at 5 mg/kg. Three mice were sacrificed at 1 hr, 24 hr, 72 hr, 168 hr and 240 hr postdose.
EDTA whole blood was collected and serum was prepared for in vivo DAR analysis by mass spectrometry.
Serum sample pre-dilution Serum samples were diluted in horse serum (Life technologies, 16050-122) based on total antibody concentrations of ADC measured by total antibody ligand binding assay. Dilutions were based on estimations of final concentration to a range of 10-30 µg/mL, which is suitable for the ic beads upper limit of binding capacity. affinity affinity purification In a protein LoBind tube (EppendorfNorth a), 350 µL horse serum was added to 100 µL of each pre-diluted ADC serum sample to a total volume of 450 µL, followed by addition of 4 µg of biotin-anti-human Fe antibody (2 µL of biotin-anti human at 2 mg/mL solution). Samples were incubated for 2 hours (hr) at room temperature by shaking at 900 rpm on an orbital shaker. For each serum sample, 50 µL slurry of streptavidin coated magnetic beads (Pierce, Cat# 88817) was equilibrated with 0.1 % Tween in PBS buffer (PBST) in a LoBind tube. Phosphate Buffered Saline with Tween 20 (PBST) buffer was removed by a pipette after pulling the magnetic beads to the side ofthe LoBind tube by placing the LoBind tube on a magnetic rack. Serum samples after 2 hr incubation with anti-human capturing reagent were transferred to the LoBind tubes ning equilibrated magnetic beads, and incubated at room temperature for 1 hr at 900 rpm on an orbital . Serum was d after magnetic bead incubation, and the magnetic bead was washed thoroughly with 500 µL PBST (3 times) ed by 500 µL 5% MeOH in MilliQ water (3 . Magnetic bead bound ADC was released by incubating the magnetic beads with 100 µL 0.5% formic acid in 50% MeOH/MilliQ water for 15 minutes.
Reduction ofpurifiedADC Released ADC was reduced by adding 10 µL reducing reagent (l0mM TCEP freshly prepared from powder sed from Thermo Scientific, with l0mM EDTA in 2M pH7.5 Tris buffer) to 100 µL ofsample and ted at 37°C for 30 minutes.
LC/MS analysis Reduced samples (10 µL) were injected into an Agilent 6550 QTof LC/MS system through a temperature controlled (5°C) CTC autosampler. Sample elution was achieved on a Waters C-4, 3.5 µm, 300 A, 2.1 x 50 mm i.d. HPLC column. The mobile phases were: A: 0.1% formic acid in water, and B: 0.1 % formic acid in MeCN; the flow rate was 0.45 mL/min; and the column compartment was maintained at 40°C. 8] The HPLC gradient was as s: Time (min) %A %B 0 95 5 0.6 95 5 I.I 10 90 2.2 10 90 2.4 95 5 3.5 95 5 High resolution MS analysis ofreduced ADC was performed on an Agilent 6550 quadruple timeof-flight nt Technology, San Clara, CA) equipped with a Dual Agilent Jet Stream electrospray ionization (ESI) source operated in the positive ion mode. Mass spectrometer was operated in the extended dynamic range (2G Hz) mode with a MS range up to 3,200 m/z. The primary ESI source was used for LC/MS analysis, and the secondary ESI probe was used for ng calibration solution at 922.009798 m/z to achieve real time MS calibration. The mass spectrometer was calibrated on a daily basis. Typical mass errors ofanalytes ve to theoretical masses were less than ± 5 parts per million in daily operations. MS data were processed using MassHunter Qual r Build 5.0.
MSspectrum deconvolution Maximum entropy method in the MassHunter Bioconfirm software package was used to deconvolute the multiple charged ion mass spectra to derive neutral molecular weight spectra. The intensity ofthe deconvoluted peak was used to calculate DAR.
DAR value ationfrom de-convoluted MS spectrum DAR values were calculated using de-convoluted MS peak intensity based on the following equations: DAR value from light chain (LC): LC DAR = (2 xpeak intensity of LCA);((peak intensity ofLC+peak intensity of LCA )) LC and LCA are light chains with zero and one drug linker, respectively.
DAR value from heavy chain (HC): HC DAR=2x(peak intensity of HCA+2xpeak intensity of HCM+3xpeak intensity of HCMA)/(peak ity ofHC+peak intensity ofHCA+peak ity ofHCM+peak intensity ofHCAJ\J\) HC, HCA, HCJ\J\ and HCJ\J\J\ are heavy chains with zero, one, two and three drug linkers, respectively.
Total DAR =LC DAR+HC DAR Results The results are shown in Figure 2. This example demonstrates that minimal loss of drug linker is observed from steroid ADC over 7 days.
Example 78: tion of human and mouse transmembrane TNF-alpha GRE reporter cell lines In order to create a parental cell line, K562 cells were seeded onto a 6 well dish (Costar: 3516) with 2 mL of complete growth medium (RPMI,10%FBS, 1%L-glutamine, 1 % Na Pyruvate and 1 % MEM NEAA) at 500,000 cells per well for 24 hours at 37°, 5% CO2. The next day, 1.5 µg of pGL4.36[Luc2P/MMTV/Hygro] (Promega: E316), 1.5 ug pG14.75 [hRLuc/CMV] (Promega: , and 3 µl of PLUS reagent (lnvitrogen: 10964-021) were diluted into 244 uL Opti-MEM (Gibco: 31985-070) and incubated at room temperature for 15 minutes. The 6[luc2P/MMTV/Hygro] vector contains MMTV LTR (Murine Mammary Tumor Virus Long Terminal Repeat) that drives the transcription of the luciferase reporter gene luc2P in response to activation of several nuclear ors such as glucocorticoid receptor and androgen receptor. The pGL4.75[hR/uc/CMV] Vector s the luciferase reporter gene hRluc (Renilla reniformis) and is designed for high expression and d anomalous ription.
After incubation, diluted DNA solution was pre-incubated with 1:1 Lipofectamine LTX solution (Invitrogen: 94756) (13.2 µl + 256.8 µl Opti-MEM) and incubated at room temperature for 25 minutes to form DNA-Lipofectamine LTX complexes. After incubation, 500 µl of DNA-Lipofectamine complexes were added directly to the well containing cells. K562 cells were transfected for 24 hours at 37°, 5% CO2 .
After incubation, cells were washed with 3 mL of PBS and selected with complete growth medium containing 125 µg/mL of hygromycin B (Invitrogen: 10687-010) for two weeks. "K562 pGL4.36[Luc2P/MMTV/Hygro]pGL4.75[hRLuc/CMV]" cells were produced.
In order to create a murine embrane TNF-alpha GRE reporter cell line, the parental cells, K562 pGL4.36[Luc2P/MMTV/Hygro]pGL4.75[hRLuc/CMV], were seeded onto 6 well dish (Costar: 3516) with 2 mL of complete growth medium (RPMI, , utamine, 1 % Na Pyruvate and 1 % MEM NEAA) at 500,000 cells per well for 24 hours at 37°, 5% CO2 . The next day, 3 µg of mFL TNFa DNA (Origene: MC208048), which encodes untagged mouse TNF, and 3 µl of PLUS reagent (lnvitrogen: 10964-021) were diluted into 244 uL Opti-MEM (Gibco: 31985-070) and incubated at room temperature for 15 minutes. After incubation, diluted DNA solution was pre-incubated with 1:1 Lipofectamine LTX solution (lnvitrogen: 94756) (13.2 uL + 256.8 uL Opti-MEM) and incubated at room temperature for 25 minutes to form DNA-Lipofectamine LTX complexes. After incubation, 500 µl of pofectamine complexes were added directly to the well ning cells. The al K562 pGL4.36[Luc2P/MMTV/Hygro]pGL4.75[hRLuc/CMV] cells were transfected for 24 hours at 37°, 5% CO2. After incubation, cells were washed with 3 mL of PBS and selected with te growth medium containing 125 µg/mL of hygromycin B (lnvitrogen: 10687-010) and 250 µg/mL G418 : 10131- 027) for two weeks. "K562 mouse FL-TNFa GRE (pGL4.36[luc2P/MMTV/Hygro])" cells were produced.
In order to create a human embrane TNF-alpha GRE reporter cell line, the parental cells, K562 pGL4.36[Luc2P/MMTV/Hygro] pGL4.75[hRLuc/CMV], were transfected with the plasmid hTNF delta 1-12 C-Myc pcDNA3. l(-) plasmid construct. This plasmid is pcDNA 3.1 (Thermofis her cat# V79020) encoding tace resistant transmembrane TNF (i.e., SEQ ID NO:1 lacking amino acids 77- 88). (See Perez C et al. Cell 63 (2): 251-8 (1990) discussing tace resistant transmembrane TNF.) These cell lines were then used in the TNF-alpha reporter assays bed in the subsequent examples.
Example 79: Activity of anti-TNF-alpha immunoconjugates in GRE transmembrane TNF-alpha reporter assays K562 parental GRE (pGL4.36[luc2P/MMTV/Hygro]) cells and K562 mFL-TNF-a or hTNF delta 1-12 GRE 36[luc2P/MMTV/Hygro]) cells were plated onto 96 well tissue culture treated white plates r: 3917) at 50,000 cells per well in 50 µL of assay medium (RPMI, 1 % CSFBS, 1 % L­ glutamine, 1 % Na Pyruvate and 1 % MEAA). The cells were treated with 25 µL of 3x serial diluted murine or human anti-TNF-a antibody drug conjugates in assay medium, steroid compound, or media alone and ted for 48 hours at 37°, 5% CO2 . After 48 hours of incubation, cells were d with 75 µL of Dual-Glo Luciferase Assay System (Promega-E2920) for 10 minutes and analyzed for luminescence using the TopCount (PerkinElmer). Data were analyzed using a four parameter curve fit to generate EC50 values. % maximum activation was normalized to l00nM dexamethasone, which was considered m activation. The results using the murine TNF-alpha cell line are shown in Table 12 below, and the results using the human TNF-alpha cell line are shown in Table 13 below. In Table 12 below, A refers to 8Cll. In Table 13 below, A refers to adalimumab (SEQ ID NOs: 66 and 73). Percent (%) monomer was determined by SEC as previously described (see ADC analytical procedures). "'d ("") 00 N 0 """ (.,; Ut Ut """ QC) :;; 0 N 0 """ � """ 0 ""­ -..l """ 46 93 K562 GRE 68 (% ax) m 74 9.3 5.27 the anti-murine K562 GRE ECso 36.3 (ug/m L} ].61 95 114 refers to mTNFa GRE( % ]06 max) 118 0.0108 0.0105 assay ( A ) GRE reporter mTNFa GREEC50 1.27 (ug/mL 0.00519 99.9 99.9 % 95.4 r 91.4 4 2 transmembrane TNFa n 4.4 4.5 - - 334 in mouse A \ A n n s-+- n " / / \ J \ SA / j ugates o H C H0 2 O \ � S ) ,c O ) N ,Jly- H drug con /', � H H O ,o,c �� o N )l___,,---.N H o _ N )l___,,---. H "�-J- O II O H � N ...._,,;--. \ � o TNFa antibody re O , O , 0 �--' o H OH, �N ...._,,;--. H Stru ctu � v::�'.j)L,-)l_, s -+-A � �N/ p 0.._('V �N )l__,, �N o Y\' --:::O .,,0 0 -y•' v J;"V 0 0 � " ,,V 0 � H ofanti-murine O o O O H I � , _ F * H o ; HO ' F HOO F \ o I � 0 * 1 ) \ 0 / \ I activity \ o 135 136 134 137 Cpd. No. Table 12: In vitro antibody 8Cl TNFa t'f, (''l (i';� r-- <r, '-0 ,'i <r, r--- I"] •n ;.:, c; r.::;i 00 <r, lr, ",;"l ,'-! ........ ;\ /\ r--°' 0\ M °' r--- "7 ....,"' ,'-! N ......., 7 r.::;i --s 0 0 0 0 0 0 0 0 00 00 00 0\ 0\ 0\ 0\ 0\ 118 95 0.0795 0.0406 0.0399 0.0393 97.7 2 4 - - 336 A A s s -+- S s tl � 0 � o 0 ) C C o � 0 N Av H _ H 0 2 N N Av H H � 2 H o,c l- J 1 o _0 H H N :(" O H , N __,r- · /·• N ,ll..___,r- O H O N� . (YY)' i- f <O :c __A \ � o o \ � 0 N � H H N :( ! � H \ '>-- �N )l_ p s ' O � O --:, 11__) \ H --t -v -·-(' o 0 H 1' , ()0 .,,0 O ·r 0--( o o H O O < * 0 *o F H O ·� � F <o Ho 0 0 � � 0 0 146 C• r--- r--- r---X X O'\ '--0 I:"- :J\ 00 C\ (") 0 r- ,::::, C-'1 ...-< ff')- 0 0'; ('·] ., ('sj 0 00 00 r--- 0\ 91 87 70 94 ] 2.1 2.85 3.44 2.77 ]04 104 96 75 04 0.0098 0.0247 0.0185 90.1 2 - - 338 A A , � i � s H ) H�C , n O 0 ) N,ll_,,,, H _ H0 2 C Ls ,:C H02C H _) ,u O , l_,,,._ H H \ 0 ,)L,,) H _ o Ho .,,N A-./'- 0 O H �,;- ¢ H N H \ O ¢ 0 O H0 --, i .f 1 0 \ � � � ()s 'O \ o H 0-- - r)l ,,10 .o''O) H \ -(' O I r<", Di O o H o o Ho I .. ,o F � F F o o i= 0 �,o aw"' 0 0 �, o-,)l � 0 153 154 151 0.2 79 ."! 7., Q'' o."! >50 2.8 15.8 3.63 0.05 94 107 115 >50 0.0171 0.039 0.0198 99.4 96 98.2 92.5 2 4 2 4 - - 339 n s )l., _ ) H0 2 C s -+- A S ) H0 2 s -+- A C o ,- - H o NY-v H _ o H) HO� .., H �C 2 � N N o 7('-" )L H o H !J- 0 II H = II O = O � O H HN-../'- N : . o N : ··•'' o 0 � H N-../'- N � N :�··•'' 0 H � N � 0 ,D- u(( -.(' v(( .••' --:: --:: ..,o 0"'0 (Y" ..,o 0,, ...- .,,0 ...,V , I .. ,o O HO O o , o.... O HO .0 o � � F i= 0 � O 0 dfW 0 � 156 157 155 158 r--. M r- C;C 0 '-0 l''· (;;C 0 0•n 0\ "'T !, ...... (''1 r- ......, 7 C', 0 C', C', 0 t', •n 0 ;\ 0 0 0 0 "'1': 00 00 tr, 0\ 0\ ""T 00 .,,.. ci ci <r;r- 0 0 0 '>I:) ",;"l <r, <n !, "T /\ ,\ 0•n 0 /\ ;\ 0 r--- 0\ 18.4 0.463 0.0108 0.0551 0.146 94.4 2 - n A - 342 ' s ' S+ '( ' f1 s ) ) l,, _ ) a a " H02C NA .,, H _ HO� NA.,, H _ 0 "':\ �� o \ l,__,,,o 0 "':\ _,,o " ? o ? \ ? H� ,, :C-) o C°'l o.._,,,, :� \ I C°'l 6 .._,/ )l} y' I " bl o bl O o 'l ) o l, a l ) �iJ- o 0 7 0 7 lf l,__,,,o l,__,,,o o l, (' • , O O i --,0 '->-: � '->-: ,.Jl,,;.-_ ,, , O I .,,0 0 Y\l v 0 o o�.- � " H 0 o 0 Y\l H i= O o O � } HO F HO F " o 0 �, - O �,. v 0 0 \10 � 170 167 __...L_ 0•n 0 /\ /\ 0 0 0 00 00 0\ 0\ <( r<rc �� Iz�o �r:ro I � IZ OJ: OJ: " o IZ ,,11u.. 55 66 48 60 >50 7.94 3.88 41.4 92 105 93 113 0.0464 8 8.00.E-04 0.0262 93.4 4 - ' - 344 H02C\ j ll s o . s a ' _ ) C H < _) H H� o )l. . .- . H0 2 )'v i ,1 30 a H cr"' -i("' )L < l,;-- o --r- !J O J) 0 �� --r-o ~ "� L! ,O j . 1 H � H �, , ("° , ..,_,, 0"'s ,O ��-f a V �--·- I 0 0"' s H ,.,,V I 0 H ..-- V 0 .. ,o .,,0 o , O o . . HO F o �, a,·" o o o H F � 0 � 0 dBP1 0 0 176 177 178 8 >50 0.0249 0.0593 0.0118 97.7 4 4 - - 345 A s , s s �'.;- o C � O H0 2 H N H02C l� 2 C l� o H O o 0 )l) � I H N j , ("" l i\ _ O J \ ii o~ : �!, f 0 " �0 , H N o v' i fl (' ..._,, j ,("" 0 0:J .-"V � 'P ..._,, 0' ' o 1 .. ,o 1 � O Jl__ __) 0-( J; " o .,10 HO i" ,o o f , 0 � 0� o o � , F ao � 0 � 0 O� _\\ 180 181 182 179 __L_ 0\ '·D '° M I:"- M r-- r-� N 0 -, lr, "T <r; ;\ 0 0 0 0•n 0 0 ",;"l ,r, /\ /\ /\ t'.'·1 r---- , 0 ......., 0 0 0 0 0 00 0\ 0\ 0\ 0\ 0\ 0\ i:--- 7" <( M (f) C <( ' o C �� '<�o z 'f (f) Cl) �0 <s IZ ���o O 0 �IZq� (s • � 0 o O 0 < IZ IZ 0 (s oJ: s ZI ZI 0 (s 0 ..... (s '"" 'Fo 'F-o IZ IZp � (s Cl) �o 0b _ � ',, 100 o �0 I 0 IZ 0 � ·- q � ••ILL � � ----;---� 0 §? "" ·••I §? � I 0 � •""- 0 \ 'f � t- 00 0\ 00..., 00..., 00..., 0\..., °' 'tj" � :J\ 0 <r; ,.....; 00 ""T M ....., 'tj" N "'1': C IC •n t'.'·l N°' <r; °' r- '·D ('·] C-'1 "'T"""' •n 0 0 0 0 0 0 0 (/)b Q d(/) ,,__!0 -:_ --. 0 . 0 0 •• Ill..
••ILL I 75 63 91 60 I 23.6 12.7 I 24.2 39.3 156 96 96 ]43 0.123 0.0331 0.0614 0.0626 I 92.2 - A - 349 ) s s 0 {l s ., OzC C H0 2 C I\ H� 2� C " Ho c s j" [ ~ I\ j-J'J ! 0~ o ~ o . J'- /'-/ 0 -../" o-J- o ~ -J-o� Q ../"() "-J"'o ~ o.J' v'D� H NY"' O H ", ("' O j '('-' o i y-J' o V s "(Y V (Y' '('( V v 0' � v _(Y v I ,,\0 (Y' ' ('( o.....,.,, ,,,0 o o, ... •V o o ,,. a , .,,0 . o o " � i= O O F 0 �, 0 � "oo 0 �H 0 196 197 198 I 73 92 98.7 70.0 I 23.3 4.76 I 50.0 50.0 I 93 114 123 116 0.0654 0.0114 0.0154 0.16 I I 90.1 99.3 99.5 93 I I 3.5 3.8 4 4 3.84 - I I A n �S-\-A - 350 � ) ) ' S I n H N H0 2 J 0 S-\-A H02C 0 ,,.,,.,... ,,.,.....__,,..� 0 � H H N Y"--' o H N '\("N °,�c 0 �� •• 0 11 " H --.:: 0 111 0 0 � .# o ,� OH HO'' \ # N � H O \1 0 S ,,( )''' � H N v ,,Q'-0 --r I: --.:: l_Dr, HO .... b s .,,0 ,,Q N ">- H / c?° ·•''vv ,,0 O - O . § .,\0 HO H N � O if ·•• 0 � � I O H 7 O • o ,\ 0 ,y N H . - 0 N� HO i= 9J I OO H � � 0 ' I A-+-S 0 ,t \� I 200 I 207 208 I I 209 199 I 97.1 146 348 I 61.7 91.7 I I 1.9 I 50.0 50.0 18.7 I 27.6 I 248 129 130 140 101 0.362 0.154 0.0341 0.0267 3 I I I 99.2 100 99.4 99.2 99.2 I I I 3.8 4 4 4.08 3.99 - I I I - 351 ) I I I ) • oe '"" � T I � " 0 ,- 0 T on•>:r L o » .,,,o OH l_ O Ii "' o O H OIi•" < ·� bH � 1 • · r o ,OJ)" 0� /� HO 4 � '' : 0 � ,,,. P \ bH 'oH # HQ ' ., HO ,..... P 0 1 1 ·"" 0 \\ 0 ,.... R ,... � .,, ,,.. HO I # HG ; 0 S J)' I � n...D " �'- '- Qo U N H 0 � 0 � H � HO Z:" O 0 , = = � O N H �� �0N 0 o 0 0 t N H --yi J 2 H 0 �J N� H 2H C 0 t "'Y'J C 0 � co,H s 0 s 0 , A l� I s __c;'" ,L H- A l 0 ,_(__ 211 212 213 214 210 103 27.9 102 112 I 97.2 I I 91.2 I 1.93 50.0 I I 18.3 I 22.9 I 50.0 I 9.96 I 126 108 101 96.0 140 97.7 I 0.0166 0.351 0.0147 0.023 0.0371 9 I I I I I 99.3 99.4 98.8 98.9 99.1 99.5 I I I I I 1.7 3.8 3.74 3.7 3.7 4.03 I - I I I I - 352 I ) I I I ) OH • · - - F o #! #! n� o bH O H 11..- r .,,,- 0 0 " r oo P bH 1 o R .,... bH o, , • . La ,,, q_ ,. . o .. P \ o u • .,... P bH ,,.. P bH \\ ,,..0 0 � HO �·-�, ' ,,,,.l . o -"" O HO ,.... J)"ol; ,,,.. Hif I .,, 0 H O ..... () # H O , HO I I -"" s J:I: �o ,, , I -.._ V S S I [ 0» -.._ � , ,YU '- '- N H 0 0 "-s. N " V O OH , N U � O - 0 0 H N� 0 �0N H H ', \,O__,O'o� O a N H lJi , O N H 0 0 a � yo\ � N H i y O ....ly CO2 H N j( H 0 0 2 H co,H HJ( co 2 H N � 0 H N � S c o , 0 , s ( � " ' '{ii(! � H L A l'( I A-\-S A J�{, / A-+-s 218 219 216 217 220 215 I 112 86.6 GRE (% max) 66 K562 4.28 the anti­ I 43.4 mL) 43 131 102 ECsO (u g/ refers to K562 GRE (% 100 assay (A 0.011 0.114 GRE max) hTNFa GRE ECso mL) (u2:/ 0.0 l 79 GRE reporter hTNFa 99 99 % monomer 97.6 and 73) 3.6 3.8 n 4 embrane TNFa NOs: 66 (SEQ ID / n in human 0 o E : ,,,,,,:;:. OH F �� " A o H OH �o L�, drug conjugates antibody adalimumab 0,,,,. 01 O ...- \ QH 1• 0 ' ' ' HO 011 o,"" O iO p \ OH --''ld· I :::,., � 0,,,,, L human TNFa ure I :::,., N� H 0 TNFa antibody � N H Stru ct 0 � J-_ , a N� H . , - O , = ', " 'J.., O H __)l N l y O H✓ �J y 0 I a N H✓ of anti-human N H 0 2H � 0 H C0 2 �J �]" _ , H N H C0 C O 0 s J-_ l ( s ---( -._)l A A In vitro activity I A--s \ 221 222 Table 13: Cpd. No. 201 103 51 35 21 I I 17.5 >50 >50 >50 I I 97 121 l] 8 103 0.0035 0.0318 0.0482 I I I 0.0767 I 96.9 99.1 98.2 99.4 2 4 I I I 2 4 I - n n - 354 c I /2 / ]__ 0 h F ;t 0 '� F F OH 'l,,. H �o HO H o_ 0 " ' � _.l_ o 0 0 OH h',, ·( . 4 � ( ,,,. 0 ' ' ' . l Ho r}� HO h .o , , Q,0 ::,,_ H H H N O'v � ,, � = 0 __)- N \ h'vl) O 0 \ .o O !J N� } --- t _)- O O NH K N \, H 0 - N H 0 O � � C0 2 H r->-- 0 AIS---{N NJN \, H C0 2 H 0 H C02H C02H s -{ O A -t- S I A---\-S-----c�� A i � 204 I 203 I 205 I 202 I Example 80: Activity of various anti-human TNF-alpha immunoconjugates in GRE transmembrane TNF­ alpha reporter assays Preparation of anti-human TNF alpha immunoconjugates 7] All proteins were conjugated to Cpd. No. 99using conditions highlighted under the general cysteine conjugation protocol in Example 36. Where indicated in Table 14 below, a ne addition (underlined) was engineered into the anti-TNF sequence to allow conjugation.
Table 14:Amino acid sequences ofanti-human TNF alpha antibodies used in conjugates Antibody Sequence (SEQ ID NO) Infliximab HC SEQ ID NO:67 imab LC SEQ ID NO:74 Golimumab HC SEQ ID NO:72 Golimumab LC SEQ ID NO:78 etanercept LPAQVAFTPYAPEPGSTCRLREYYDQTAQMCCSKCSPGQHAKVFCTKTSDT VCDSCEDSTYTQLWNWVPECLSCGSRCSSDQVETQACTREQNRICTCRPGW QEGCRLCAPLRKCRPGFGVARPGTETSDVVCKPCAPGTFSNTTSST DICRPHQICNVVAIPGNASMDAVCTSTSPTRSMAPGAVHLPQPVSTRSQHTQ PTPEPSTAPSTSFLLPMGPSPPAEGSTGDEPKSCDKTHTCPPCPAPELLGGPSV PKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKP REEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT PPVLDS DGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:79) ABT-122HC SGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWVSA ITWNSGHIDYADSVEGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAKVSY LSTASSLDYWGQGTLVTVSSGGGGSGGGGSEVQLVQSGAEVKKPGSSVKV SCKASGGSFGGYGIGWVRQAPGQGLEWMGGITPFFGFADYAQKFQGRVTIT ADESTTTAYMELSGLTSDDTAVYYCARDPNEFWNGYYSTHDFDSWGQGTT VTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALT SGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVE PKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE DPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKE YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVK GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:80) ABT-122LC DIQMTQSPSSLSASVGDRVTITCRASQGIRNYLAWYQQKPGKAPKLLIYAAS TLQSGVPSRFSGSGSGTDFTLTISSLQPEDVATYYCQRYNRAPYTFGQGTKV EIKRGGSGGGGSGEIVLTQSPDFQSVTPKEKVTITCRASQDIGSELHWYQQKP DQPPKLLIKYASHSTSGVPSRFSGSGSGTDFTLTINGLEAEDAGTYYCHQTDS LPYTFGPGTKVDIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKV QWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEY THQGLSSPVTKSFNRGEC (SEQ ID NO:81) certolizumab SEQ ID NO:68 pegol HC certolizumab SEQ ID NO:75 pegol LC izumab HC EVQLVESGGGLVQPGGSLRLSCAASGYVFTDYGMNWVRQAPGKGLEWMG WINTYIGEPIYADSVKGRFTFSLDTSKSTAYLQMNSLRAEDTAVYYCARGY RSYAMDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNV NHKPSNTKVDKKVEPKSCDKTHTCAAHHHHHH (SEQ ID N0:82) izumab LC DIQMTQSPSSLSASVGDRVTITCKASQNVGTNV A WYQQKPGKAPKALIYSA SFLYSGVPYRFSGSGSGTDFTLTISSLQPEDFA TYYCQQYNIYPLTFGQGTKV EIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC (SEQ ID N0:83) Adalimumab Fab EVQLVESGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWVSA HC ITWNSGHIDYADSVEGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAKVSY LSTASSLDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNV NHKPSNTKVDKKVEPKSCDKTHTCAAHHHHHH (SEQ ID N0:84) Adalimumab Fab DIQMTQSPSSLSASVGDRVTITCRASQGIRNYLAWYQQKPGKAPKLLIYAAS LC TLQSGVPSRFSGSGSGTDFTLTISSLQPEDVATYYCQRYNRAPYTFGQGTKV EIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC (SEQ ID N0:85) dy1 GVDNKFNKENIAAMTEITRLPNLNPYQRAAFIWSLSDDPSQSANLLAEAKKL NDAQAPKC (SEQ ID N0:86) Ozoralizumab EVQLVESGGGLVQPGGSLRLSCAASGFTFSDYWMYWVRQAPGKGLEWVSE Nanobody INTNGLITKYPDSVKGRFTISRDNAKNTLYLQMNSLRPEDTAVYYCARSPSG FNRGQGTLVTVSSGGGGSGGGSEVQLVESGGGLVQPGNSLRLSCAASGFTF SSFGMSWVRQAPGKGLEWVSSISGSGSDTLYADSVKGRFTISRDNAKTTLY LQMNSLRPEDTAVYYCTIGGSLSRSSQGTLVTVSSGGGGSGGGSEVQLVESG GGLVQPGGSLRLSCAASGFTFSDYWMYWVRQAPGKGLEWVSEINTNGLIT KYPDSVKGRFTISRDNAKNTLYLQMNSLRPEDTAVYYCARSPSGFNRGQGT LVTVSSGSEQKLISEEDLCHHHHHH (SEQ ID N0:87) 1 Kronqv1st Jet al. Protem Engmeenng, Design & Select10n 21 (4).247-255 (2008) ty ofanti-human TNF alpha immunoconjugates in GRE reporter assay Anti-human TNF alpha immunoconjugates (also referred to as anti-human TNF alpha ADCs, or anti-hTNF alpha steroid ADCs) were tested for activity in the K562 parental GRE (pGL4.36[luc2P/MMTV/hydgro]) and K562 hTNF delta 1-12 GRE (pGL4.36[luc2P/MMTV/hydgro]) cell lines under conditions as described in Example 79. The results shown in Table 15 te that all antihTNF alpha d ADCs tested demonstrate potent antigen-dependent activity dissociated from their activity on the parental cell line.
Table 15: In vitro activity of anti-human TNF alpha ADCs in human transmembrane TNFa GRE reporter assay (ADC concentrations were normalized for MW and DAR) HN_)- '--::: '--::: K\ vv,,, OH 0 r>-NH o '( O .,,H H HO 0 H .,,F��i \ C02H hTNFa hTNFa K562 K562 Cpd. % GRE GRE GRE GRE Al SEQ ID n No. r EC50( (% EC50 (% nM) max) (nM) max) 233 infliximab 67/74 3.3 99.99 1.3 122 374 55.3 234 mab 72/78 4.9 99 3.3 144 1633 67.5 235 etanercept 79 2.3 87.6 0.6 105.5 164 104 236 ABT-122 80/81 4.1 99.98 1.02 116.9 608 61.3 certolizumab 237 68/75 2 95.9 0.44 97 1111 39 pegol 238 certolizumab 82/83 1 ND 0.4 95 266 60 adalimumab 239 84/85 l ND 1 118 190 137 240 affibody 86 1 100 9.1 98 26 84 ozoralizumab 241 87 1 98 0.5 131 875 99 (nanobody) Binding of anti-human TNF alpha conjugates to human TNFalpha Binding kinetics of anti-h TNF alpha steroid ADCs to recombinant soluble TNFa trimer were ined by surface plasmon resonance-based measurements made on Biacore T200 instrument (GE Healthcare) at 25°C using either anti-human Fe/ anti-human F(ab'h e (used for all ADCs except affibody and ozoralizumab ADCs) or direct NHS/EDC mediated amine coupling ch (used only for ozoralizumab ADC). Approximately 10000 RU of goat anti-human IgG Fe polyclonal antibody (Thermo Fisher Scientific Inc., cat. No. 31125) or goat anti-human F(ab'h polyclonal antibody (Jackson Immunoresearch Laboratories, Inc. cat. No. 109006) was diluted to 5 µg/mL in 10 mM sodium acetate (pH 4.5) and was immobilized across a CMS biosensor chip using a standard amine coupling kit according to manufacturer's instructions and procedures. Unreacted es on the biosensor surface were blocked with IM lamine. For direct amine coupling approach, approximately 750 RU of ozoralizumab steroid conjugate was directly immobilized onto CMS chip. Chip preparation and binding c measurements were made in the assay buffer HBS-EP+ (10 mM Hepes, pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.05% Tween 20). For binding kinetic measurements in capture format, each assay cycle consisted of the following steps: 1) capture of test ADC on test surface at a concentration of 0.5 µg/mL and at a flow rate of 5 µL/min for 60 s; 2) analyte injection (human TNFa or buffer only) over both nce and test surface for 300s at 50 , after which the dissociation was monitored for 600 seconds at 50 µ1/min; 3) regeneration ofcapture e by 10 mM Glycine-HCl, pH 1.5 or 100 mM HCl (for directly coupled ADC) injections over both reference and test surface. For binding kinetics measurements in direct amine coupling format, only step 2) and step 3) were performed. During the assay, all measurements were referenced against the blank surface alone (i.e. with no captured test antibody or immobilized nanobody) and buffer-only injections were used for double referencing. TNFa injections ranged in concentration from 50 nM to 0.39 nM in a 2-fold dilution series, respectively. Data were processed and fitted globally to a 1:1 binding model using Biacore T200 Evaluation software to determine the binding kinetic rate constants, ka (M 1s 1) and kct (s 1), and the brium dissociation constant KD (M).
Two independent experiments were conducted. Reported values in Table 16 are es from these experiments.
Table 16: Binding affinities of TNF alpha steroid ADCs to human TNF alpha (ND not determined) anti-TNF Cpd No. ka (1/Ms) kct (lls) KD(M) 233 infliximab 9.90E+05 3.l0E-04 10 234 golimumab 9.70E+05 2.40E-04 2.50E-10 235 etanercept 6.30E+06 9.80E-05 l.60E-ll 236 ABT-122 4.00E+05 6.90E-05 l.80E-10 certolizumab 237 2.30E+06 04 7.30E-ll pegol 238 certolizumab 2.70E+06 l.60E-04 6.00E-11 adalimumab 239 l.50E+06 l.80E-04 l.30E-10 240 affibody ND ND ND ozoralizumab 241 l.30E+06 7.00E-05 5.20E-ll (nanobody) Example 81: Activity ofanti-hTNF alpha steroid ADCs in Lipopolysacharride Stimulated Human PBMC Cytokine Release Assay Primary human eral blood mononuclear cells (PBMCs) were purchased from Biological Specialty Corporation (cat# 21410), washed in 50 mL PBS, re-suspended in FBS with 5% DMSO, aliquoted and cryopreserved in liquid nitrogen until use. The PBMCs were thawed, re-suspended in RPMI supplemented with 2% FBS, and 1 % Penicillin-Streptomycin, and plated into a cell assay plate (Costar #3799). The cells were incubated with varying concentration of TNF alpha steroid ADCs at 37 °C and 5% CO2 for 4 hours. Cells were then stimulated with l00ng/ml LPS for overnight. On the following day, plate was spun for five s at 1000 rpm, and 100 µL of supernatant media was directly transferred to an additional l plate and analyzed for IL-6 (MSD, #Kl51AKB) and IL-1 beta(MSD, GB) concentrations. The dose response data were fitted to a sigmoidal curve using nonlinear regression, and the IC50 values calculated with the aid of GraphPad 5.0 (GraphPad Software, Inc.). The results shown in Table 17 trate that the anti-hTNF alpha steroid ADCs have potent activity in inhibiting the release of pro-inflammatory nes IL-6 and IL-lbeta from activated primary immune cells.
Table 17: In vitro activity of anti-human TNF alpha ADCs in LPS-stimulated human PBMC cytokine release assay (n 2) Cytokine Release IC50 (ng/ml) Cpd No.
IL-6 IL-lbeta 203 14.3 ± 3.5 3.6 ± 1.2 201 86.8 ± 69.6 25.5 ± 21.3 205 42.4 ± 27.9 22 ± 18 Example 82: Activity of anti-TNF-alpha immunoconjugate in TNFa-induced cytotoxicity assay in L929 cells L929 is a murine aneuploid fibrosarcoma cell line that is sensitized by pretreatment with mycin D. Treatment with TNFa initiates apoptosis and subsequent cell death. L929 cells in log phase were ted using trypsin 0.05%, washed twice with D-PBS and counted by CEDEX. The cells were resuspended at 1E6 cell/mL in assay media containing 4 µg/mL mycin D and 50 µL was added to all wells. Anti-murine TNF alpha steroid ADC (anti-murine TNF alpha 8Cl 1 conjugated to Cpd 71; also referred to as anti-mTNF alpha steroid ADC) and urine TNF mAb (8Cl 1) were diluted to a 4x concentration in assay media and serial 1:3 dilutions were performed. Mouse TNFa was diluted to a 4x concentration of 600 pg/mL. The anti-mTNF steroid ADC and anti-mTNF mAb (125 µL) were added to the mTNFa (125 µL) in a 1:2 dilution scheme and allowed to te for 1 hour at room temperature, gently shaking. The antibody/mTNFa (or ADC/ mTNFa) mixture was added to wells at 50 µL/well in cate. The plates were incubated for 20 hours at 37 °C, 5 % CO2 . To quantify viability, 10 µL of WST-1 reagent (Roche cat# 11644807001) was added to wells. Plates were incubated under assay conditions for 3.5 hours, centrifuged at 500 xg and 75 µL supernatant transferred to an ELISA plate (Costar cat#3369). The plates were read at OD 420-600 nm using a Spectromax 190 ELISA plate reader.
Data was analyzed and IC50 values calculated using a sigmoidal dose response ble slope) fit in GraphPad Prism 5.
Anti-mTNF alpha steroid ADC had comparable neutralizing potency (IC50 l.9nM) to unconjugated anti-mTNF alpha mAb (IC50 l.5nM).
Anti-human TNF alpha immunoconjugates were tested for neutralizing activity under conditions described above. The results are shown in Table 18 and indicate the anti-human TNF alpha immunoconjugates tested demonstrate potent neutralization ofhuman TNF alpha.
Table 18: lization potencies of anti-hTNF alpha steroid ADCs to human TNF alpha-induced cytoxicity in L929 cells (ND= not ined) SEQ ADC % IC50 Compound anti-TNF ID DAR monomer (nM) 233 imab 67/74 3.3 99.99 ND 234 golimumab 72/78 4.9 99 0.050 235 etanercept 79 2.3 87.6 0.002 236 ABT-122 80/81 4.1 99.98 0.074 certolizumab 237 68/75 2 95.9 0.046 pegol 238 certolizumab 82/83 1 ND 0.085 adalimumab 239 84/85 1 ND 0.6 240 affibody 86 l 100 ND ozoralizumab 241 87 l 98 0.018 (nanobody) Example 83: Binding of anti-mTNF-alpha d ADC to mouse Fcgamma receptors SPR (surface plasmon resonance) based Biacore T200 instrument (GE Healthcare) was used to evaluate binding of TNF-alpha steroid ADC (anti-mTNF 8Cl 1 conjugated to Cpd 71) and antimTNF-alpha mAb to recombinant mouse FcgRs (all R&D s). The FcgRs were directly immobilized on the surface of the flow cells two, three and/or four of the CMS type S Biacore chip(s) to e densities of 2000 RU (resonance units). Blank modified surface of the flow cell one of each Biacore chip was used as a reference surface. Each experiment consisted of association and dissociation phases. Association phase consisted of titrating al mAb and ADC over all flow cells at a flow rate of 50 ul/min and concentrations of 4000, 2000, 1000, 500, 250, 125, 62.5, 31.25 and 0 nM for FcgRIIB and FcgRIII and 100, 50, 25, 12.5, 6.25, 3.13, 1.56 and O nM for receptors I and IV. Dissociation phase consisted of the continuous flow of the running buffer (HBS-EP+, pH 7.4, GE Healthcare)at the flow rate of 50 ul/min. Association and iation phases were monitored for 5 min each (receptors I and IV) or 1 min (receptors II and III). Chip es were regenerated with a 5s pulse of100 mM HCl at a flow rate of 100 ul/min after each binding cycle. Biacore Evaluation software was used to fit the raw data to 1:1 (FcgRI and IV) or Steady State (receptors IIB and III) binding models. Results are shown in Table 19. ka is the association rate constant (1/Ms); kct is the dissociation rate constant (1/s); KD is the equilibrium dissociation constant (M).
Table 19: Binding ties of anti-TNF-alpha immunoconjugate to mouse a receptors muFcgRI muFcgRIIb muFcgRIII muFcgRIV ka (1/Ms) kct (lls) KD (M) KD (M) KD (M) ka (1/Ms) kct (lls) KD (M) Anti-mTNF 7.4E+05 l.6E-02 2.lE-08 3.7E-06 3.lE-06 5 5.3E-03 4.6E-08 Anti-mTNF 3.2E+05 8.7E-03 8 5.9E-06 6 8.4E+04 4.6E-03 5.5E-08 Example 84: Activity of anti mTNF-alpha steroid ADCs in contact hypersensitivity model Anti-mTNF alpha steroid ADCs were evaluated in an acute contact hypersensitivity model, an elicitation of acute skin inflammation using delayed-type ensitivity (DTH) response (T-cell driven) via application of a sensitizing agent (fluorescein isothiocyanate (FITC)). The cy of anti-mTNF alpha steroid ADCs was measured by the ability to reduce ear swelling. The steroid biomarkers corticosterone and procollagen type 1 N-terminal propeptide (PlNP) were included as readouts to assess the putative impact of anti-mTNF alpha steroid ADC treatment on the Hypothalamus-Pituitary-Adrenal (HPA) axis and bone turnover respectively.
Ear Swelling On day O mice were placed under general anesthesia and the abdomens were . Using a ipettor, mice were sensitized by epicutaneous applicaton of400uL of FITC solution (1.5% on in 1:1 acetone:DBP) on the abdomen. 6 days later mice were dosed with vehicle or therapeutic agent 1 hour prior to ear challenge with FITC. For ear challenge, mice were placed under general anesthesia and were challenged with 20µ1 FITC applied onto right ear. 24 hours after nge mice were placed under general esia and their ear thickness is measured by r. Difference between challenged and unchallenged ears was calculated. 72 hours after ear challenge, mice were injected with ACTH at lmpk IP, and terminally bled at 30min post-ACTH. Plasma is collected and analyzed PlNP, corticosterone, free steroid, and large molecule levels.
Quantification of edfree steroid and endogenous corticosterone Calibration curve of steroid was prepared in mouse plasma with final concentrations from 0.03 nM to 0.1 µM at 8 different tration levels. Corticosterone calibration curve ranging from 0.3 nM to 1 µM final corticosterone concentrations was prepared in 70 mg/mL bovine serum albumin solution in PBS buffer. A solution of 160 µL MeCN with 0.1 % formic acid was added to 40 µL study plasma samples or calibration standards. Supernatants were diluted with distilled water and 30 µL final sample solution was injected for LC/MS analysis.
Quantification of released free steroid and osterone was conducted on an AB Sciex 5500 triple quadruple mass spectrometer connected to a Shimadzu AC20 HPLC system interfaced with an electrospray ionization source ing in positive mode. A Waters e BEH Cl8, 2.lx30mm, 3.5 µm column was used for tography separation. The mobile phase A was 0.1 % formic acid in Milli Q HPLC water, and mobile phase B was 0.1 % formic acid in MeCN.A linear gradient from 2% ofmobile phase B to 98% mobile phase B was applied from 0.6 to 1.2 minutes. The total run time was 2.6min at a flow rate of 0.8 mL/min. The mass spectrometer was operated in positive MRM mode at source temperature of 700°C.
Quantification ofplasma P JNP 9] Quantification of plasma PlNP was conducted on a LC/MS platform based on protein trypsin digestion. Plasma samples were partially precipitated and fully reduced by adding MeCN/0.lM ammonium bicarbonate/OTT mixture. Supernatant was collected and alkylated by adding iodoacetic acid.
The alkylated proteins were digested by trypsin and ing tryptic peptides were analyzed by LC/MS. ation curve were generated by using synthetic tryptic peptide spiked into horse serum (noninterfering surrogate matrix). Stable isotope d flanking peptide (3-6 amino acids extension on both termini of the tryptic peptide) was used as internal standard added in the MeCN/DTT protein precipitation mixture to normalize both ion ency and LC/MS injection.
A Columnex Chromenta BB-Cl8, 2.lxl50mm, 5 µm column was used for chromatography separation. The mobile phase A was 0.1 % formic acid in Milli Q HPLC water and mobile phase B was 0.1 % formic acid in MeCN. A linear gradient from 2% of mobile phase B to 65% mobile phase B was d from 0.6 to 3 min. The total run time was 8min at a flow rate of 0.45 mL/min. An AB Sciex 4000Qtrap mass spectrometer was used in positive MRM mode to quantify PlNP peptides, at source temperature of 700°C.
Quantification oftotal ADC in plasma Concentrations of total antibody (ADC and backbone mAb) were measured by ligand binding assay using Mesoscale Discovery (MSD) platform. Biotin d mouse TNF was used as the capture reagent for anti-mTNF alpha steroid ADCs and Sulfa-TAG conjugated goat anti-mouse detection antibody was used for detection. A calibration curve was generated by serial on of the ADC molecule in matching matrix and QC samples were used to qualify the assay Results The s are shown in Table 20 below: Table 20: Comparison ofanti-mTNF alpha steroid ADC activity on ear swelling and steroid biomarkers in CHS model of inflammation swelling PlNP ADC (% inhib SEM (% inhib.@ SEM Corticosterone(% SEM lOmpk) inhib@ lOmpk) lOmpk) Cpd. No. 151 87.8 3.5 32.3 3.9 71.5 5.6 Cpd. No. 145 87.8 3.4 19.2 6.3 15.1 9.9 Cpd. No. 169 90.2 2.2 38.3 2.8 60.1 4.7 Cpd. No. 167 86.1 2.4 26.1 6.9 48.3 5.1 Cpd. No. 162 76.3 2.7 25.9 6.5 50.4 5.5 Cpd. No. 161 64.4 4.6 1.4 7.2 37.1 4.1 Cpd. No. 172 79.8 3.9 14.6 4.9 6.3 6.8 Cpd. No. 176 81.4 3.9 20.0 7.9 15.0 9.4 Cpd. No. 177 94.3 1.2 27.0 6.0 17.1 9.0 Cpd. No. 180 80.8 2.1 45.7 6.0 39.9 3.8 Cpd. No. 149 92.4 2.3 52.8 2.8 74.4 3.7 Cpd. No. 175 66.5 4.7 12.1 4.5 49.3 3.5 Cpd. No. 207 87.3 3.6 44.0 5.4 54.6 5.2 Cpd. No. 178 94.4 1.8 58.0 2.4 73.8 4.7 Cpd. No. 181 78.8 4.6 -13.2 7.2 29.4 8.0 Cpd. No. 182 60.1 5.6 -15.1 11.5 3.4 4.8 Cpd. No. 185 85.0 4.0 51.6 7.0 43.9 9.6 Cpd. No. 186 70.5 3.9 1.5 9.4 19.1 3.9 These results demonstrate that TNF alpha steroid ADCs can obtain an cious response equivalent to small molecule steroid treatment while sparing the undesired effects on corticosterone and PlNP.
An additional contact hypersensitivity (CHS) study was conducted to address whether conjugation of the steroid payload to anti-TNF mAb was required for enhanced efficacy. Mice were dosed i.p. once according to the protocol described above with either vehicle, anti-mTNF alpha mAb (l0mpk), anti-mTNF alpha steroid ADC (l0mpk) (cpd no 139) or a mixture of anti-mTNF alpha mAb co-dosed (concurrently delivered in a single i.p. injection) with an equivalent amount of small molecule steroid to match the ADC iometry. For a l0mpk dose of anti-mTNF alpha steroid ADC with a DAR of 4, this was calculated to be 4 µg of small molecule steroid (Cpd. No. 42). The results shown in Figure 9 demonstrate that anti-mTNF alpha steroid ADC treatment had significantly increased cy in reducing ear inflammation when compared to the combination of anti-mTNF alpha mAb and small molecule steroid or anti-mTNF alpha mAb alone.
Example 85: Activity of anti-mTNF-alpha steroid ADCs in collagen-induced tis The ability of TNF alpha steroid ADC (Cpd. No. 137) to impact disease was assessed in the collagen-induced arthritis (CIA) model of arthritis.
In these experiments, male DBA/lJ mice were obtained from Jackson Labs (Bar Harbor, ME). Mice were used at 6 to 12 weeks of age. All animals were maintained at nt temperature and humidity under a 12-hour light/dark cycle and fed with rodent chow (Lab Diet 5010 PharmaServ, Framingham, MA) and water ad libitum. AbbVie is AAALAC (Association for Assessment and Accreditation of Laboratory Animal Care) accredited, and all procedures were approved by the Institutional Animal Care and Use Committee (IACUC) and monitored by an attending narian. Body weight and condition were monitored, and animals were euthanized if exhibiting >20% weight loss.
The male DBA/J mice were immunized intradermally (i.d.) at the base of the tail with 100 µL of emulsion containing 100 µg of type II bovine collagen (MD ences) dissolved in 0.1 N acetic acid and 200 µg of nactivated Mycobacterium tuberculosis H37Ra (Complete Freund's Adjuvant, Difeo, Laurence, KS). Twenty-one days after immunization with collagen, mice were d IP with 1 mg of n A , St. Louis, MO) in PBS. Following the boost, mice were monitored 3 to 5 times per week for arthritis. Rear paws were evaluated for paw swelling using Dyer spring calipers (Dyer 5) 8] Mice were ed between days 24 and 28 at the first clinical signs of disease and buted into groups of equivalent arthritic severity. Early therapeutic treatment began at the time of enrollment.
Animals were dosed once orally (p.o.) with steroid (Cpd. No. 3) (l0mpk) in a 0.5%HPMC/0.02%Tween80 vehicle [v/] or intraperitoneal (i.p.) with anti-mTNF alpha mAb (l0mpk) (8Cl1) or anti-mTNF alpha steroid ADC (l0mpk) (Cpd. No. 137) in 0.9% saline. Blood was collected for antibody exposure by tail nick at 24 and 72 hours after dose. Paws were collected at the al timepoint for histopathology. Blood was ted at the terminal timepoint by cardiac puncture for complete blood counts (Sysmex 0iV). Statistical significance was ined by ANOVA.
The results are shown in Figure 3 and demonstrate that a single dose of anti-mTNF alpha d ADC can exhibit an extended duration of action through amelioration of paw swelling for -6 weeks compared to anti-mTNF alpha mAb or small le steroid alone.
In a separate study designed to address the TNF-targeting functionality of the anti-mTNF alpha steroid ADC, animals were dosed once i.p. with anti-mTNF alpha mAb (l0mpk) or TNF alpha steroid ADC (l0mpk) (Cpd. No. 145) or with isotype steroid ADC (l0mpk) (Cpd. No. 224): Lo,·· 0 J 1 �JOJ)'''' A N N,.,..r( : N H H = H OH Cpd. No. 224 n A= Anti-OVAL (Gallus gallus) which recognizes the hen egg protein ovalbumin, an antigen not expressed in mice. Both ADCs had lent drug load. Small molecule steroid (3 mpk) was dosed orally once daily (q .d). The results are shown in Figure 10 and demonstrate that a single dose of anti-mTNF alpha steroid ADC has equivalent cy to small molecule steroid dosed daily over a 21 day period. A single dose of the rgeted isotype steroid ADC had only partial efficacy, similar to anti-mTNF mAb alone over the same timeframe.
Percentages denote % inhibition compared to vehicle. An evaluation of the animals' body weights throughout the course of this study (Figure 11) revealed all the treatment groups with the exception of the anti-mTNF alpha steroid ADC group lost weight. In contrast, the anti-mTNF alpha steroid ADC treated mice exhibited normal weight gain throughout the 21 day study.
Example 86: ty of various anti-mTNF alpha steroid ADCs in collagen-induced arthritis 2] Several anti-mTNF alpha steroid ADCs with different steroid payloads or drug:antibody ratios (DARs) were tested for efficacy in a mouse model of arthritis. The studies were conducted according to the ure outlined in Example 85. The results are shown in Table 21 below.
Table 21: Efficay ofanti-mTNF alpha steroid ADCs in model of arthritis '% inhibition ofpaw swelling vs Cpd No. DAR(n) SEM vehicle (AUC0-21d) (at lOmpk) 136 2 75 3.7 137 4 91 6.5 139 4 93 2.9 143 2 96 3.3 145 4 95 4 151 4 101 2.5 172 4 74 9.3 176 4 85 8.3 177 4 99 4.2 Example 87: Activity ofanti-hTNF-alpha conjugates in human TNF transgenic Tgl278TNF knock-out mouse model ofCollagen Antibody Induced Arthritis The efficacy of anti-human TNF alpha ADCs was assessed in a human TNFa transgenic mouse model of arthritis.
The Collagen Antibody-Induced Arthritis (CAIA) model (Moore, AR J Transl Med 12:285 (2014)) was performed using the human TNF enic Tgl278TNF knock-out mice as previously described (Moore A et al. J Transl Med 12 (1): 285 (2014)). Eight mgs of a il of monoclonal antibodies that target different epitopes of collagen type II (ArthritoMab™) were administered intraperitoneally (i.p.) to the mice on day 0. On day 3, the mice were ed i.p. with 10 µg LPS to boost the disease pathology. Animals were evaluated for tic score daily starting from day 3 until day 14 of the study. Eight male mice were used per group and test articles or PBS vehicle were administered i.p. twice a week for two weeks.
The results are shown in Figure 4 and demonstrate that anti-human TNF alpha ADCs can icantly reduce disease score ed to an anti-human TNF alpha mAb (adalimumab).
Example 88: Activity ofanti-mTNF alpha steroid ADCs on peak inflammation A mouse CIA experiment was conducted to establish the efficacy of anti-mTNF alpha steroid ADC on animals with peak inflammation. For late therapeutic dosing, mice were enrolled in the study at first clinical signs of disease and dosed 6 days after enrollment. A group of s was sacrificed at day 7 of disease to e a baseline for arthritic changes by micro-computed tomography (µCT) and histologic analysis at the time all other groups were dosed. All animals were dosed once on day 6 with either vehicle (0.9% saline), anti-mTNF alpha mAb (l0mpk) (8Cll) or anti-mTNF alpha steroid ADC (l0mpk) (Cpd. No. 145) and sacrificed on day 21. Arthritic hind paws were collected and µCT analysis was performed. The same paws were then used for ogic evaluation. At the termination of the experiment, whole blood was collected by cardiac puncture to evaluate complete blood counts (CBCs).
Micro-Computed Tomography ( µCT) Rear paws were removed intact at the tibia/fibula and fixed in 10% Formalin. Paws were scanned by µCT o Medical AG, Micro-CT40) at 55 kVp at 145 µA at High Resolution setting (1000 Projections/180° at 2048x2048 Pixel Reconstruction) using Isotropic Voxels and 300 millisecond integration time. A cylindrical contour was manually drawn around region of interest from the tibiotalar junction and extending into the ankle for 100 slices (1.8 mm). Evaluation was performed by Scanco software utilizing 0.8 sigma gauss, with an upper threshold of 1000 and a lower threshold of 320. ogic evaluation 8] Rear paws from treated mice were immersion fixed in 10% neutral buffered formalin and partially decalcified in Calrite solution for 48 hours to allow trimming of the lateral and medial edges of the tarsus. Paws were then placed back into Calrite for -48 hours to complete decalcification. Samples were routinely processed, embedded in paraffin in the sagittal plane, ned at 5 microns and stained with hematoxylin and eosin. Slides were evaluated copically for the presence of inflammation/ pannus formation, phil infiltration, bone erosion and cartilage damage using a 0-4 scale: 0= none t, 1 = mild, 2 = moderate, 3 = marked, 4 = severe.
The results shown in Figure 12 demonstrate that a single dose of anti-mTNF alpha steroid ADC can reverse established disease and reduce paw swelling to near baseline. In contrast, a single dose ofanti-mTNF alpha mAb had a minimal effect on inflammation.
The effect of treatment on tarsal bone loss as measured by µCT is shown in Figure 13. The results demonstrate that a single dose of anti-mTNF alpha steroid ADC administered at peak of mation is able to significantly inhibit disease-mediated joint bone erosion compared to anti-mTNF alpha mAb alone.
The results of histological evaluation of the joints of d CIA mice are shown in Figures 14-17. They demonstrate that a single dose of anti-mTNF alpha steroid ADC administered at peak disease resulted in a significant decrease in inflammation, pannus formation, bone erosion and cartilage damage by day 21 relative to age-matched vehicle controls (p<0.001), and levels of disease were equivalent to the levels observed in controls at baseline (vehicle d6). In two of six paws evaluated, no e was detectable in the / phalangeal joints of TNF alpha steroid ADC treated animals at day 21, as compared to 100% incidence in mice at day 6 baseline (prior to treatment) and day 21 vehicle treated mice.
In contrast, a single dose of anti-mTNF alpha mAb at peak disease did not inhibit inflammation, bone erosion, pannus formation, or cartilage ction, relative to age-matched vehicle controls at d2l. Levels of e were more severe than baseline ls, and a mild trend for improved inflammation was observed.
Whole blood was ed to te changes m peripheral blood cell subsets with treatment. The results shown in Figure 18-23 demonstrate that the increase in some peripheral blood cell tions observed in diseased s can be resolved with a single dose of anti-mTNF alpha steroid ADC. Statistically significant reductions in overall white blood cells, neutrophils and monocytes were observed with anti-mTNF alpha steroid ADC treatment.
Example 89: Comparison of anti-mTNF-alpha steroid ADCs and anti-CD163 ADCs To demonstrate the enhanced eutic efficacy of an anti-TNF immunoconjugate in the treatment of inflammatory disease, we compared its activity to an ADC targeting the hemoglobin scavenger receptor CD163, a glucocorticoid immunoconjugate approach described in the ture to have targeted anti-inflammatory functionality (PCT Int. Appl. 039510A2 by Graversen NJH, et al.; Graversen JH et al., Mal. Ther. 20 (8): 1550-8 ).
Generation of a mouse CDI63 GRE report cell line A parental cell line was created similar to that described in Example 78 but with CHO-K l cells instead of K562 cells. The resulting parental cell line CHO pGL4.36[Luc2P/MMTV/Hygro] PGL4.75[hRLuc/CMV] was then transfected with a plasmid which encodes mouse CD163 (Origene cat.no. MR216798) under conditions described in Example 78. The resulting cell line CHO mCD163 GRE (pGL4.36[luc2P/MMTV/Hygro]) was used to test the in vitro activity of both anti-mTNF-alpha and ouse CD163 immunoconjugates (also referred to as antimCD163 immunoconjugates or anti-mCD163 steroid ADC).
Preparation of an anti-mouse CD163 immunoconjugate A chimeric rat anti-mouse CD163 mlgG2a/k antibody was generated from the VH and VL ce for clone 3El0B10 as described (SEQ ID NO:87/88 from PCT Int. Appl. WO201l/039510A2).
This antibody was conjugated to Cpd. No. 99 using conditions highlighted under the general cysteine conjugation protocol in Example 36 to give a drug:antibody ratio (DAR) of 4.
Activity of an anti-mouse CDI63 conjuate in mouse CDI63 GRE reporter assay The anti-mouse CD163 immunoconjugate was tested for activity on the CHO mCD163 GRE (pGL4.36[luc2P/MMTV/Hygro]) cell line under conditions described in e 79. An anti-mTNF alpha steroid ADC (Cpd. No. 145) was included as a negative control. The results in Table 22 demonstrate the anti-mouse CD163 immunoconjugate (Cpd. No. 223): Lo••·· o J l �J�,--· A N N'l( : N OH H H = H Cpd. No. 223 A= anti-CD163 shows antigen-dependent activity dissociated from the anti-mTNF alpha d ADC on the mouse CD163 GRE cell line.
Table 22 mCD163 mCD163 CHOGRE CHO Cpd No. DAR GREEC50 GRE(o/o EC50 o monomer (ug/ml) max) (ug/ml) max) 223 4 93 0.2 92 >50 55 145 4 97 >20 70 >50 39 Activity ofanti-mouse CDI63 conjugate in mouse collagen-induced arthritis The y of anti-mouse CD163 d immunoconjugate to impact paw swelling was assessed in the en-induced arthritis (CIA) model of RA. A control anti-mTNF alpha d ADC (cpd 139) with the same drug-linker and DAR as the CD163 steroid ADC was also evaluated in the same study and the parental mAbs for both ADCs were also included as treatment groups. The experiment was conducted according to the procedure outlined in Example 85. The s are shown in Figure 24 and demonstrate that while the CD163 steroid ADC initially reduces paw swelling in the first few days after single dose treatment, this effect is transient. In comparison, a single dose of anti-mTNF alpha steroid ADC is sufficient to completely suppress inflammation through the duration of thestudy It is to be appreciated that the Detailed Description section, and not the Summary and Abstract sections, is intended to be used to interpret the claims. The Summary and Abstract sections sets forth one or more, but not all, exemplary embodiments of the present disclosure as plated by the inventor(s), and thus, are not intended to limit the present disclosure and the appended claims in any way.
The present disclosure has been described above with the aid of functional building blocks illustrating the implementation of specified functions and relationships thereof. The boundaries of these functional building blocks have been arbitrarily defined herein for the convenience of the description.
Alternate boundaries can be defined so long as the specified functions and relationships thereof are appropriately performed.
The foregoing description of the ic embodiments will so fully reveal the general nature of the disclosure that others can, by applying knowledge within the skill of the art, readily modify and/or adapt for vanous ations such specific embodiments, without undue experimentation, without departing from the general concept of the present sure. Therefore, such adaptations and modifications are intended to be within the meaning and range of equivalents of the sed embodiments, based on the ng and guidance presented herein. It is to be understood that the phraseology or terminology herein is for the purpose of description and not of limitation, such that the terminology or phraseology of the present specification is to be interpreted by the skilled artisan in light of the teachings and guidance.
The breadth and scope of the present sure should not be limited by any of the abovedescribed exemplary embodiments, but should be defined only in accordance with the following claims and their equivalents.

Claims (27)

WHAT IS CLAIMED IS:
1. A compound having Formula I-a: (SM-L-Q)n -A1 I-a wherein: A1 is an anti -tumor necrosis factor (TNF) alpha protein; Lis a ; Q is a heterobifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a monovalent l of a glucocorticosteroid.
2. A compound having a 1-b: (SM-L-Q)u -A2 1-b A2 is a protein; Lis a linker; Q is a heterobifunctional group or heterotrifunctional group; or Q is absent; n is 1-10; and SM is a radical of a glucocorticosteroid represented by Formula 11-m or Formula 11-p: R6a R6a 3 z x*y2 R3 R ,..- 1/ Z X R6b R9f I I � ......__.., R9f 1/ S' I *I � .,,o-1.. ,,g, ,,,,:; 7 6e R6c .,,o-J.) ,-y 6e ,,,,;;; y2"\ ·•10 R11b R '''0 R�R R6d R6d 0 11-p 11-m 0 wherein: R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; -373 - R3 is selected from the group consisting of -CH20H, -CH2SH, -CH2Cl, -SCH2Cl, -SCH2F, - SCH2CF3, hydroxy, -0CH2CN, -0CH2Cl, -0CH2F, -0CH3, -0CH2CH3, -SCH2CN, /�o ": OH 0 .,'OH '-.//, .0 R3b /....._'-./ n.O R3c x;x Y n0 CO2H R3a O ' O ' and R3a is selected from the group consisting of hydrogen and C1.4 alkyl; R3b is selected from the group consisting of C1.4 alkyl and C1.4 alkoxy; R3c is selected from the group consisting of hydrogen, C1.4 alkyl, -CH20H, and C1.4 alkoxy; R3d and R3e are ndently selected from hydrogen and C1.4 alkyl; R6a, R6\ R6C, R6d, and R6e are each independently selected from the group consisting of hydrogen, halo, C1.4 alkyl, C1.4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and ; X is selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=0)-, -S(=0)r, -NR5-, - CH2S-, -CH20-, -N(H)C(R8a)(R8b)-, CR4d-, and -C=C-; or Xis absent; Y2 is selected from the group consisting of , -S-, and -N(R7a)-; or Y2 is absent; tis I or 2; Z is selected from the group consisting of =CR11a- and =N-; each R4a and R4b are independently selected from the group ting ofhydrogen and C1.4 alkyl; R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4d are independently selected from the group consisting of hydrogen and C1.4 alkyl; R5 is selected from the group consisting of hydrogen and C1.4 alkyl; R7a is selected from the group ting of hydrogen and C1.4 alkyl; R8a and R8b are independently selected from the group ting of hydrogen and C1.4 alkyl; R9f is ed from the group consisting of hydrogen and C1.4 alkyl; R11a and R11b are independently selected from the group consisting of hydrogen, halo, C1.4 alkyl, C1.4 haloalkyl, cyano, hydroxy, thiol, amino, hio, and alkoxy; and =-= represents a single or double bond.
3. The compound of claim 1 or 2, wherein SM 1s a radical of a glucocorticosteroid represented by Formula 11-m: (Jz x R6a R3 y� R9f I ;q I s ' .,,o-J·•'' r' ✓,;:. 6e R6c ••10 R11b R 11-m; R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group consisting of , -CH2SH, -CH2Cl, l, - SCH2F, -SCH2CF3, hydroxy, -0CH2CN, -0CH2Cl, -0CH2F, -0CH3, -0CH2CH3, N, ./-...........,..o �OH y·,,oH l-...........,..o O R3b / R3c Y -...........,.. o C02H R3a O'l( 'l( ' O ' and R3a is selected from the group consisting of hydrogen and C1.4 alkyl; R3b is selected from the group consisting ofC1.4 alkyl and C1.4 alkoxy; R3c is selected from the group consisting of hydrogen, C1.4 alkyl, , and C1.4 alkoxy; R3d and R3e are independently selected from hydrogen and C1.4 alkyl; R6a, R6c, R6d, and R6e are each independently selected from the group consisting of hydrogen, halo, C1.4 alkyl, C1.4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=0)-, -S(=0)2-, - NR\ -CH2S-, -CH20-, -N(H)C(R8a)(R8b)-, -CR4c=CR4d-, and -C=C-; or Xis absent; Y2 is ed from the group consisting of , -S-, and -N(R7a)-; or Y2 is absent; tis I or 2; Z is CH-; each R4a and R4b are independently selected from the group consisting of hydrogen and C1.4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4d are independently selected from the group ting of hydrogen and C1.4 alkyl; R5 is selected from the group ting of hydrogen and C1.4 alkyl; R7a is selected from the group consisting of hydrogen and C1.4 alkyl; R8a and R8b are independently selected from the group ting of hydrogen and C1.4 alkyl; R9f is selected from the group consisting of hydrogen and C1.4 alkyl; R11b is selected from the group consisting of hydrogen, halo, C1.4 alkyl, C1.4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
4. The compound of claim 2 or 3, wherein: =-= ents a double bond; R is selected from the group consisting of hydrogen and fluoro; R2 is selected from the group consisting of en and fluoro; R3 is selected from the group consisting of -CH2OH, -CH2Cl, -SCH2Cl, -SCH2F, and /'-.._,,,,0, /? �-o-R3d O-R3e R3d and R3e are independently selected from the group consisting of hydrogen, methyl, and ethyl; R6a, R6C, R6d, and R6e are hydrogen;X is selected from the group consisting of -CH2-, , -S-, -S( O)-, -S( O)r, -CH2S-, and -N(H)CHr; Y2 is -N(H)-; Z is CH-; R9f is hydrogen; and R11b is hydrogen.
5. The compound of any one of claims 1-4, wherein Lis a linker comprising a dipeptide.
6. The nd of any one of claims 1-5, wherein Q is a heterobifunctional group selected from the group consisting of: \�Ns-l HO C 0 H .._ and 11 N � s/'l.. 0 0 Q-3 Q-4 and mis 1, 2, 3, or 4.
7. The compound of any one of claims 1-6, wherein-L-Q-is: 10b HO C H � 0 H � \. \ 'l(' ¼,N S/ R10a O H 0 m is 2 or 3; and R10a and R10b are independently selected from the group ting of hydrogen and C1.4 alkyl.
8. The compound of any one of claims 1-7, wherein n is 2-5.
9. The compound of claim 1 or 2, wherein SM is a monovalent radical of a glucocorticosteroid which is any one of the compounds of Table 11.
10. The compound of any one of claims 1 or 3-8, wherein A1 is (i) an antibody or antigenbinding fragment thereof that binds to human TNFalpha or (ii) a soluble TNF receptor.
11. The nd of any one of claims 1 or 3-9, wherein A1 is selected from the group consisting of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab.
12. The compound of claim 1, which is any one or more of the nds of Table III, wherein: n is 1-5; AisA1; and A1 is selected from the group consisting of adalimumab, infliximab, certolizumab pegol, afelimomab, nerelimomab, ozoralizumab, placulumab, and golimumab.
13. The compound of claim 2, which is any one or more of the compounds of Table III, wherein: n is 1-5; AisA2;and A2 is an antibody or a soluble receptor protein.
14. Acompound selected from the group consisting of: •''ld NJN\, �JN S H H = H O - OH •''ld NJN\, �JN S-{ H H = H O - OH 0 \ �N-( � 0 '=- V,,,.(o �NH 0 � 0 A sJN \_ H 0 -le/O" O O O ,,, A l S--c� �\N'I � OH A s�NJ)y�)-N�" l:H H H = H wherein n is 1-5 and A is an antibody comprising the heavy and light chain sequences of SEQ ID N0:66 and SEQ ID N0:73, respectively.
15. The compound of claim 14 selected from the group consisting of: Structure n A 2 o ,Q_,() J'o'· 0 2 A s-<:: N�N�MJ I : N :::,.. :::,.. H H O = H OH 0 o A s -- ( :"�"�M J () lcfH : N :::,.. OH O = H C0 2H
16. The compound of claim 15, wherein the compound is Structure n A 4
17. The compound of claim 15, wherein the nd is Structure n
18. The compound of claim 15, wherein the compound is Structure n
19. The compound of claim 15, wherein the compound is Structure n
20. The compound of claim 15, wherein the compound is ure n .l ,-· ,,. 4 O O O 0 A s-C� ,,____)l � ,r"'I'� OH
21. The compound of claim 15, wherein the compound is Structure n s-C� ll!-� .l ,-· ,,____)l ,,. 2 O O O 0 A OH
22. A pharmaceutical ition comprising the compound of any one of claims 1-21, and a pharmaceutically acceptable carrier.
23. A method for treating an autoimmune disease in a patient in need f comprising administering to said patient the compound of any one of claims 1-21 or the pharmaceutical composition of claims 22, optionally wherein said autoimmune disease is rheumatoid arthritis, juvenile idiopathic arthritis, tic arthritis, ankylosing spondylitis, adult Crohn's disease, ric Crohn's disease, ulcerative colitis, plaque psoriasis, hidradenitis suppurativa, uveitis, Behcets disease, a loarthropathy, or psoriasis.
24. A nd having Formula VII: or a pharmaceutically able salt or solvate thereof, wherein: R1 is selected from the group consisting ofhydrogen and halo; R2 is selected from the group consisting ofhydrogen, halo, and hydroxy; R3 is selected from the group consisting of -CH20H, -CH2SH, -CH2Cl, -SCH2Cl, , -SCH2CF3, -CH20S(=0)20H, hydroxy, -OCH2CN, -OCH2Cl, -OCH2F, -OCH3, -OCH2CH3, - /'--...,.,,-o yy oH o y •,, o H I ,.,,-O O R 3b /'--...,.,,-0n R 3c C02H R3a O ' O ' and R3a is selected from the group consisting of hydrogen and C1.4 alkyl; R3b is selected from the group consisting of C1.4 alkyl and C1.4 alkoxy; R3c is ed from the group consisting of hydrogen, C1.4 alkyl, -CH20H, C1.4 alkoxy, -CH2(amino), and -CH2CH2C( =O)OR3r; R3d and R3e are independently selected from the group consisting of hydrogen and C1.4 alkyl; R3f is selected from the group consisting of hydrogen and C1.4 alkyl; Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -S(=O)-, -S(=0)2-, - NR\ -CH2S-, -CH20-, (R8a)(R8b)-, -CR4c=CR4d-, -C=C-, -N(R5)C(=O)-, and -OC(=O)-; Xis absent; tis I or 2; -383 - Z is selected from the group consisting of=CR11a- and =N-; each R4a and R4b are ndently selected from the group consisting of hydrogen and C1.4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4d are ndently selected from the group consisting of hydrogen and C1.4 alkyl; R5 is selected from the group ting of hydrogen and C1•4 alkyl; R6a, R6\ R6C, and R6d are each independently selected from the group ting of hydrogen, halo, C1•4 alkyl, haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; R7a is selected from the group consisting of hydrogen and C1.4 alkyl; R7b is selected from the group consisting of hydrogen, -L-H, -L-PG, 0 0 �Q L-N> O , and O ; or R7a and R7b taken together with the nitrogen atom to which they are attached form: O ; or R7a and R7b taken together with the nitrogen atom to which they are attached form a nitro group; mis 1, 2, 3, 4, 5, or 6; Lis a linker; PG is a protecting group; R9f is selected from the group consisting of hydrogen and C1.4 alkyl; R8a and R8b are independently selected from the group consisting of hydrogen and C1.4 alkyl; R11a and R11b are independently selected from the group consisting of en, halo, C1•4 alkyl, Cn haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
25. A compound having Formula VII-A or Formula VII-B: 0 6b R3w9f,,, z I x� r-.t 7b ,o -�N R7a '''0 R11b R60 VII-A or R2 VII-B, or a pharmaceutically acceptable salt or solvate thereof, wherein: R1 is selected from the group consisting of hydrogen and halo; R2 is selected from the group consisting of hydrogen, halo, and hydroxy; R3 is selected from the group consisting of-CH20H, -CH2SH, -CH2Cl, -SCH2Cl, -SCH2F, -SCH2CF3, -CH20S(=0)20H, hydroxy, -OCH2CN, -OCH2Cl, -OCH2F, -OCH3, -OCH2CH3, /�o�OH oy•,, 3b /�o oH /�oYOnR R3c C02H R3a O ' O ' and R3a is selected from the group consisting of hydrogen and C1.4 alkyl; R3b is selected from the group consisting of C1.4 alkyl and C1.4 alkoxy; R3c is selected from the group consisting of hydrogen, C1.4 alkyl, -CH20H, C1.4 alkoxy, -CH2(amino), and -CH2CH2C(=O)OR3r; R3d and R3e are independently ed from the group consisting of hydrogen and C1.4 alkyl; R3f is selected from the group consisting of hydrogen and C1.4 alkyl;Xis selected from the group consisting of -(CR4aR4b)t-, , -S-, -, -S(=O)r, -NR5-, -CH2S-, -CH20-, (R8a)(R8b)-, -CR4c=CR4d-, -C=C-, -N(R5)C(=O)-, and -OC(=O)-; or Xis absent; tis I or 2; Z is ed from the group consisting of=CR11a- and =N-; each R4a and R4b are independently selected from the group ting of hydrogen and C1.4 alkyl; or R4a and R4b taken together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl; R4c and R4d are independently selected from the group consisting of hydrogen and C1.4 alkyl; R5 is selected from the group consisting of hydrogen and C1.4 alkyl; R6a, R6\ and R6c are each independently selected from the group consisting of hydrogen, halo, C1•4 alkyl, haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; R7a is selected from the group consisting of hydrogen and C1.4 alkyl; R7b is selected from the group consisting of hydrogen, -L-H, -L-PG, j-L-N� and O ; or R7a and R7b taken together with the nitrogen atom to which they are attached form: O ; or R7a and R7b taken together with the nitrogen atom to which they are attached form a nitro group; mis 1, 2, 3, 4, 5, or 6; Lis a linker; PG is a ting group; R9f is selected from the group consisting of hydrogen and C1.4 alkyl; R8a and R8b are independently selected from the group consisting of hydrogen and C1.4 alkyl; R11a and R11b are independently selected from the group ting of hydrogen, halo, C1•4 alkyl, C1-4 haloalkyl, cyano, hydroxy, thiol, amino, alkylthio, and alkoxy; and =-= represents a single or double bond.
26. The compound of claims 24 or 25, or a pharmaceutically acceptable salt or solvate thereof, n: R7b is ed from the group consisting of: , and mis 1, 2, 3, 4, 5, or 6; and R10a and R10b are each independently selected from the group consisting of hydrogen and optionally substituted C1-6 alkyl.
27. The compound of claims 24 or 26, or a pharmaceutically able salt or solvate thereof, having Formula VIII-a: R6a R7b R3 JZ C X¢N'R?a R9f I I ,,102.J,,,, ":1,
NZ788703A 2016-06-02 2017-06-01 Glucocorticoid receptor agonist and immunoconjugates thereof NZ788703A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US62/344,948 2016-06-02
US62/371,134 2016-08-04

Publications (1)

Publication Number Publication Date
NZ788703A true NZ788703A (en) 2022-05-27

Family

ID=

Similar Documents

Publication Publication Date Title
KR102435599B1 (en) Glucocorticoid receptor agonists and immunoconjugates thereof
CN110291097A (en) Steroidal compounds and its protein-conjugate
EP3658192B1 (en) Glucocorticoid receptor agonist and immunoconjugates thereof
TW202304996A (en) Anti-b7-h3 antibodies and antibody drug conjugates
JP2024508081A (en) Biologically active substance conjugates, their preparation methods and their uses
MX2015003903A (en) Antibody-drug conjugate.
WO2019106608A1 (en) Anti-cd40 antibody drug conjugates
EP4289857A1 (en) Drug conjugate of glucocorticoid receptor agonist, and application thereof in medicine
WO2023025248A1 (en) Steroid compound and conjugate thereof
WO2023040793A1 (en) Anti-inflammatory compound and use thereof
NZ788703A (en) Glucocorticoid receptor agonist and immunoconjugates thereof
TW202327619A (en) Steroidal compound and conjugate thereof
RU2776489C2 (en) Antibody-drug conjugate
CA3220081A1 (en) Protein-antiviral compound conjugates