Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(11): 7280-7303, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37040336

ABSTRACT

Herein, we describe the identification, chemical optimization, and preclinical characterization of novel soluble guanylate cyclase (sGC) stimulators. Given the very broad therapeutic opportunities for sGC stimulators, new tailored molecules for distinct indications with specific pharmacokinetics, tissue distribution, and physicochemical properties will be required in the future. Here, we report the ultrahigh-throughput (uHTS)-based discovery of a new class of sGC stimulators from an imidazo[1,2-a]pyridine lead series. Through the extensive and staggered optimization of the initial screening hit, liabilities such as potency, metabolic stability, permeation, and solubility could be substantially improved in parallel. These efforts resulted ultimately in the discovery of the new sGC stimulators 22 and 28. It turned out that BAY 1165747 (BAY-747, 28) could be an ideal treatment alternative for patients with hypertension, especially those not responding to standard anti-hypertensive therapy (resistant hypertension). BAY-747 (28) demonstrated sustained hemodynamic effects up to 24 h in phase 1 studies.


Subject(s)
Guanylate Cyclase , Hypertension , Humans , Soluble Guanylyl Cyclase/metabolism , Guanylate Cyclase/metabolism , Hypertension/drug therapy , Vasodilator Agents , Pyridines/pharmacology , Pyridines/therapeutic use , Nitric Oxide/metabolism
2.
J Med Chem ; 63(15): 8025-8042, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32338514

ABSTRACT

Inhibition of monopolar spindle 1 (MPS1) kinase represents a novel approach to cancer treatment: instead of arresting the cell cycle in tumor cells, cells are driven into mitosis irrespective of DNA damage and unattached/misattached chromosomes, resulting in aneuploidy and cell death. Starting points for our optimization efforts with the goal to identify MPS1 inhibitors were two HTS hits from the distinct chemical series "triazolopyridines" and "imidazopyrazines". The major initial issue of the triazolopyridine series was the moderate potency of the HTS hits. The imidazopyrazine series displayed more than 10-fold higher potencies; however, in the early project phase, this series suffered from poor metabolic stability. Here, we outline the evolution of the two hit series to clinical candidates BAY 1161909 and BAY 1217389 and reveal how both clinical candidates bind to the ATP site of MPS1 kinase, while addressing different pockets utilizing different binding interactions, along with their synthesis and preclinical characterization in selected in vivo efficacy models.


Subject(s)
Antineoplastic Agents/metabolism , Cell Cycle Proteins/metabolism , Drug Delivery Systems/methods , Drug Discovery/methods , M Phase Cell Cycle Checkpoints/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Spindle Apparatus/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Dogs , Female , HT29 Cells , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints/physiology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Structure, Tertiary , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Rats, Wistar , Spindle Apparatus/metabolism , Treatment Outcome
3.
ChemMedChem ; 13(5): 437-445, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29451369

ABSTRACT

Rogaratinib (BAY 1163877) is a highly potent and selective small-molecule pan-fibroblast growth factor receptor (FGFR) inhibitor (FGFR1-4) for oral application currently being investigated in phase 1 clinical trials for the treatment of cancer. In this publication, we report its discovery by de novo structure-based design and medicinal chemistry optimization together with its pharmacokinetic profile.


Subject(s)
Drug Discovery , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Thiophenes/pharmacology , Humans , Models, Molecular , Molecular Structure , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Pyrroles/chemistry , Small Molecule Libraries/chemistry , Thiophenes/chemistry
4.
J Med Chem ; 60(12): 5146-5161, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28557445

ABSTRACT

The first-in-class soluble guanylate cyclase (sGC) stimulator riociguat was recently introduced as a novel treatment option for pulmonary hypertension. Despite its outstanding pharmacological profile, application of riociguat in other cardiovascular indications is limited by its short half-life, necessitating a three times daily dosing regimen. In our efforts to further optimize the compound class, we have uncovered interesting structure-activity relationships and were able to decrease oxidative metabolism significantly. These studies resulting in the discovery of once daily sGC stimulator vericiguat (compound 24, BAY 1021189), currently in phase 3 trials for chronic heart failure, are now reported.


Subject(s)
Heart Failure/drug therapy , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Soluble Guanylyl Cyclase/metabolism , Structure-Activity Relationship , Administration, Intravenous , Administration, Oral , Animals , Blood Pressure/drug effects , Chemistry Techniques, Synthetic , Dogs , Hepatocytes/drug effects , Heterocyclic Compounds, 2-Ring/administration & dosage , Humans , Male , NG-Nitroarginine Methyl Ester/adverse effects , Pyrimidines/administration & dosage , Rats, Transgenic , Rats, Wistar , Soluble Guanylyl Cyclase/genetics
5.
Mol Cancer Ther ; 16(5): 893-904, 2017 05.
Article in English | MEDLINE | ID: mdl-28292941

ABSTRACT

C4.4A (LYPD3) has been identified as a cancer- and metastasis-associated internalizing cell surface protein that is expressed in non-small cell lung cancer (NSCLC), with particularly high prevalence in the squamous cell carcinoma (SCC) subtype. With the exception of skin keratinocytes and esophageal endothelial cells, C4.4A expression is scarce in normal tissues, presenting an opportunity to selectively treat cancers with a C4.4A-directed antibody-drug conjugate (ADC). We have generated BAY 1129980 (C4.4A-ADC), an ADC consisting of a fully human C4.4A-targeting mAb conjugated to a novel, highly potent derivative of the microtubule-disrupting cytotoxic drug auristatin via a noncleavable alkyl hydrazide linker. In vitro, C4.4A-ADC demonstrated potent antiproliferative efficacy in cell lines endogenously expressing C4.4A and inhibited proliferation of C4.4A-transfected A549 lung cancer cells showing selectivity compared with a nontargeted control ADC. In vivo, C4.4A-ADC was efficacious in human NSCLC cell line (NCI-H292 and NCI-H322) and patient-derived xenograft (PDX) models (Lu7064, Lu7126, Lu7433, and Lu7466). C4.4A expression level correlated with in vivo efficacy, the most responsive being the models with C4.4A expression in over 50% of the cells. In the NCI-H292 NSCLC model, C4.4A-ADC demonstrated equal or superior efficacy compared to cisplatin, paclitaxel, and vinorelbine. Furthermore, an additive antitumor efficacy in combination with cisplatin was observed. Finally, a repeated dosing with C4.4A-ADC was well tolerated without changing the sensitivity to the treatment. Taken together, C4.4A-ADC is a promising therapeutic candidate for the treatment of NSCLC and other cancers expressing C4.4A. A phase I study (NCT02134197) with the C4.4A-ADC BAY 1129980 is currently ongoing. Mol Cancer Ther; 16(5); 893-904. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Adhesion Molecules/immunology , Immunoconjugates/administration & dosage , Aminobenzoates/chemistry , Aminobenzoates/immunology , Animals , Antibodies, Monoclonal/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/immunology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/immunology , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Mice , Oligopeptides/chemistry , Oligopeptides/immunology , Paclitaxel/administration & dosage , Paclitaxel/immunology , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinblastine/immunology , Vinorelbine , Xenograft Model Antitumor Assays
6.
Cancer Res ; 76(21): 6331-6339, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27543601

ABSTRACT

The fibroblast growth factor receptor FGFR2 is overexpressed in a variety of solid tumors, including breast, gastric, and ovarian tumors, where it offers a potential therapeutic target. In this study, we present evidence of the preclinical efficacy of BAY 1187982, a novel antibody-drug conjugate (ADC). It consists of a fully human FGFR2 monoclonal antibody (mAb BAY 1179470), which binds to the FGFR2 isoforms FGFR2-IIIb and FGFR2-IIIc, conjugated through a noncleavable linker to a novel derivative of the microtubule-disrupting cytotoxic drug auristatin (FGFR2-ADC). In FGFR2-expressing cancer cell lines, this FGFR2-ADC exhibited potency in the low nanomolar to subnanomolar range and was more than 100-fold selective against FGFR2-negative cell lines. High expression levels of FGFR2 in cells correlated with efficient internalization, efficacy, and cytotoxic effects in vitro Pharmacokinetic analyses in mice bearing FGFR2-positive NCI-H716 tumors indicated that the toxophore metabolite of FGFR2-ADC was enriched more than 30-fold in tumors compared with healthy tissues. Efficacy studies demonstrated that FGFR2-ADC treatment leads to a significant tumor growth inhibition or tumor regression of cell line-based or patient-derived xenograft models of human gastric or breast cancer. Furthermore, FGFR2 amplification or mRNA overexpression predicted high efficacy in both of these types of in vivo model systems. Taken together, our results strongly support the clinical evaluation of BAY 1187982 in cancer patients and a phase I study (NCT02368951) has been initiated. Cancer Res; 76(21); 6331-9. ©2016 AACR.


Subject(s)
Aminobenzoates/therapeutic use , Antibodies, Monoclonal/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Oligopeptides/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/analysis , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Receptor, Fibroblast Growth Factor, Type 2/immunology , Xenograft Model Antitumor Assays
7.
ChemMedChem ; 8(7): 1067-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23671017

ABSTRACT

Lead optimization of a high-throughput screening hit led to the rapid identification of aminopyrimidine ZK 304709, a multitargeted CDK and VEGF-R inhibitor that displayed a promising preclinical profile. Nevertheless, ZK 304709 failed in phase I studies due to dose-limited absorption and high inter-patient variability, which was attributed to limited aqueous solubility and off-target activity against carbonic anhydrases. Further lead optimization efforts to address the off-target activity profile finally resulted in the introduction of a sulfoximine group, which is still a rather unusual approach in medicinal chemistry. However, the sulfoximine series of compounds quickly revealed very interesting properties, culminating in the identification of the nanomolar pan-CDK inhibitor BAY 1000394, which is currently being investigated in phase I clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Discovery , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Sulfoxides/pharmacology , Uterine Cervical Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Female , HeLa Cells , High-Throughput Screening Assays , Humans , Mice , Models, Molecular , Molecular Structure , Molecular Weight , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/administration & dosage , Pyrimidines/chemical synthesis , Rats , Structure-Activity Relationship , Sulfoxides/administration & dosage , Sulfoxides/chemical synthesis , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/metabolism
8.
ChemMedChem ; 2(1): 63-77, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17131463

ABSTRACT

X-ray structures from CDK2-aminopyrimidine inhibitor complexes led to the idea to stabilize the active conformation of aminopyrimidine inhibitors by incorporating the recognition site into a macrocyclic framework. A modular synthesis approach that relies on a new late-stage macrocyclization protocol that enables fast and efficient synthesis of macrocyclic aminopyrimidines was developed. A set of structurally diverse derivatives was prepared. Macrocyclic aminopyrimidines were shown to be multitarget inhibitors of CDK1/2 and VEGF-RTKs. In addition, potent antiproliferative activities toward various human tumor cells and a human tumor xenograft model were demonstrated.


Subject(s)
Amines/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Design , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Amines/chemical synthesis , Cell Line, Tumor , Humans , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
11.
Fortschr Chem Org Naturst ; 87: 1-80, 2004.
Article in English | MEDLINE | ID: mdl-15079895

ABSTRACT

Starting off in the early 90's the field of cephalostatin analogues has continually expanded over the last 10 years. First syntheses prepared symmetric analogues like 14b (119) and 26 (65), which were subsequently desymmetrized to provide analogues like beta-hydroxy ketone 31 (19). Importantly the straightforward approach provided already compounds with mu-molar potency and the same pattern of activity as cephalostatin 1 (1) (see Chapter 2.1). Chemically more demanding, two new methods for the directed synthesis of (bissteroidal) pyrazines were devised and subsequently applied to a wide variety of differently functionalized coupling partners. These new methods allowed for the synthesis of various analogues (Chapter 2.2.; and, last but not least, for the totals synthesis of several cephalostatin natural products; Chapter 1.). Functionalization and derivatization of the 12-position was performed (Chapter 2.1 and 3) and synthetic approaches to establish the D-ring double bond were successfully investigated (Chapter 3). [figure: see text] Dealing synthetically with the spiroketal moiety, novel oxidative opening procedures on monomeric delta 14, 15-steroids were devised as well as intensive studies regarding spiroketal synthesis and spiroketal rearrangements were conducted (Chapter 3.2. and 4.). Last but not least direct chemical modification of ritterazines and cephalostatins were studied, which provided a limited number of ritterazine analogues (Chapter 4.). All these synthetic activities towards analogues are summarized in Fig. 18. During this period of time the growing number of cephalostatins and ritterazines on the one hand and of analogues on the other hand provided several SAR trends, which can guide future analogue synthesis. The combined SAR findings are displayed in Fig. 19. So far it is apparent that: Additional methoxylations or hydroxylations in the steroidal A ring core structure (1-position) are slightly decreasing activity (compare cephalostatin 1 1 to cephalostatins 18, 19, 10, and 11). Not investigated by preparation of analogues. Additional hydroxylations in the B-ring (7- and 9-position) do not have a strong effect. They appear to decrease slightly the activity in the case of 9-position (compare cephalostatin 1 1 to cephalostatin 4) and are neutral in the case of the 7-position (compare ritterazines J and K). Analogue synthesis confirmed this: 7-ring-hydroxylation has little impact on activity, e.g. 109a (Table 6). C'-ring aryl compounds with a 12,17 connected spiroketal area are much less active (cephalostatins 5 and 6), meaning South 6 moiety reduces activity [figure: see text] Confirmed by analogue synthesis, e.g. 190a and 190b (Table 9). Regarding 12-functionalization it is apparent, that all cephalostatins/ritterazines possess either a free hydroxy or a keto function at this position (exemption: cephalostatins 5 and 6--very low activity). However, it is not apparent whether a 12,12'-diol or a 12-keto-12'-ol is favored. In the cephalostatin series the most potent compounds possess a 12-keto-12'-ol function, while in the ritterazine series the direct comparison of ritterazine B and ritterazine H clearly favors the 12,12'-diol setting. Synthesis of simple analogues like 31 showed a "cephalostatin trend" for favoring the 12-keto, 12'-alcohol functionalization. Synthesis of a cephalostatin 1-12'-alcohol 1a supported that trend (2 fold drop in activity). Synthesis of acylated ritterazine B derivatives proved that free hydroxy groups in 12-position are necessary for high activity. At least one 14,15-double bond is part of all highly active cephalostatins/ritterazines. All ritterazines lacking this feature display only low potency (but most of them possess the unfavorable North A moiety or have unfavorable combinations of moieties; vide infra). However, the 14,15-double bond may be necessary "only" for stereochemical reasons creating a specific "curvature" of the molecule by "bending" the D-ring down (for an in depth discussion on this topic: see Chapter 3). In line with this are the observations that 14,15-alpha-epoxides do substantially decrease activity (cephalostatins 14 and 15) while a 14,15-beta-epoxide does not decrease activity (cephalostatin 4). Also in line with the "curvature theory" is the fact that ritterazine B (14-beta-hydrogen) is even more potent than ritterazine G (14,15-double bond). Therefore it is not clear if--at least one--14,15-double bond is essential for high activity. The synthesis and biological evaluation of completely 14-beta-saturated analogues (like 14'-beta-hydrogen ritterazine B) could answer this question. Synthesis of the partially saturated analogues 14' alpha-cephalostatin 1 1c and 7-deoxy-14' alpha-ritterazine B 2a showed that the stronger the divergence of conformation implied by the saturation is, the higher is the loss of activity, thus underlining the "curvature hypothesis". Synthesis showed, that analogues possessing the 14,15-double bond(s) are substantially better soluble, e.g. 26. Furthermore, the D-Ring area turned out to be sensitive for modifications, since substantially differing analogues, like 162, 163, and 164 were completely inactive. At least one 17-hydroxy group is part of all highly active cephalostatins/ritterazines. Loss of one out of two 17-hydroxy groups does not decrease activity (compare ritterazine K and L) but of the second 17-hydroxy groups (along with the 7-hydroxy group) as seen in the ritterazine series (compare ritterazines A/T and B/Y) leads to a significant decrease in activity. Increased activity of 17-ether analogues 178 and 179 points into the same direction All highly active cephalostatins and ritterazines are substantially asymmetric. Cephalostatins and ritterazines that are symmetric--either consisting of two polar units (cephalostatin 12 and ritterazine K) or two unpolar units (ritterazine N and ritterazine R)--or almost symmetric (cephalostatin 13 and ritterazine J, L, M, O, S) show substantially diminished potency. However, one has to keep in mind, that even some of the symmetrical compounds (e.g. ritterazine K--96 nM in the NCI panel) still show strong cytostatic properties. Same trend was identified with simple analogues, e.g. compare 26 to 31. In addition to the basic requirement of overall substantial asymmetry for high activity there appears to be the necessity for a "polarity match" between both steroidal units (33)--as one has to be substantially more polar (high hydroxylation grade) than the other. (e.g. cephalostatin 1 (1): North 1--high hydroxylation grade--and South 1--low hydroxylation grade; or: ritterazine B (2): South 7--medium hydroxylation grade--and North G--very low hydroxylation grade). Not directly confirmed by Analogue Synthesis--some "polarity matched analogues" did not show appropriate activity, e.g. 198 and 197. 4 core moieties are privileged, meaning all highly active ritterazines/cephalostatins (see table 1) are constructed out of them. Namely these are North 1, South 1, South 7 and North G. Numerous analogues were prepared to probe questions regarding the mechanism of action of the cephalostatins, e.g. close cephalostatin analogues like 197 and 198 (70) with increased energy content in the spiroketal. However, so far the mechanism and mode of action of the cephalostatins remains unknown. In the absence of any structural information of the biological target(s), the understanding about the structural necessities for high cytostatic activity is still limited and thus the rational design of more simple, yet highly active analogues seems at the current stage elusive. Additionally, there are many open questions, e.g. how the "monomeric" OSW-1 (3) relates to the "dimeric" cephalostatins. It remains the hope that forthcoming studies will bring light into this so far nebulous area--enabling chemists in the long run to provide highly active analogues in substantial amounts for advanced pharmacological studies. In conclusion one can state that the first decade after the extraordinarily complex cephalostatin 1 (1) entered the scene was necessary for the chemists to explore novel ways towards cephalostatins and cephalostatin analogues. They have provided methods to prepare basically every thinkable cephalostatin analogue, have delivered simple analogues (< 10 steps) with substantial activity and shaped first SAR trends in the class of cephalostatins. Now the time has come for chemists to harvest the fruits of their long and enduring synthetic ventures by aiming towards highly active, yet still not too complex analogues, which could be available in substantial amounts for advanced pharmacological studies. And for pharmacologists to explore the therapeutic potential of the cephalostatins along with elucidation of the unknown mechanism. Clearly, there is much more to expect of the cephalostatins in the coming years.


Subject(s)
Antineoplastic Agents/chemistry , Phenazines/chemistry , Spiro Compounds/chemistry , Alkadienes/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Biological Factors/chemistry , Biological Factors/metabolism , Cell Line, Tumor , Methods , Molecular Structure , Phenazines/chemical synthesis , Phenazines/metabolism , Spiro Compounds/chemical synthesis , Spiro Compounds/metabolism , Stereoisomerism , Steroids/chemistry , Structure-Activity Relationship
12.
Angew Chem Int Ed Engl ; 38(4): 549-552, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-29711784

ABSTRACT

A seven-step cascade reaction-in which selective mesylation, epoxide formation, epoxide lysis, cyclization, reiterative oxidation, and nitrogen-oxygen exchange occur sequentially-facilitates the construction of the maleic anhydride moiety of CP molecules 1 and 2 (>93% yield per step). Unstable intermediates of this reaction sequence were detected, providing evidence for the proposed mechanism and resulting in the discovery of a new chemical entity.

SELECTION OF CITATIONS
SEARCH DETAIL
...