Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 42
Filtrer
1.
Mol Cancer Ther ; 20(8): 1367-1377, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34045230

RÉSUMÉ

Targeted, catalytic degradation of oncoproteins using heterobifunctional small molecules is an attractive modality, particularly for hematologic malignancies, which are often initiated by aberrant transcription factors and are challenging to drug with inhibitors. BRD4, a member of the bromodomain and extraterminal family, is a core transcriptional and epigenetic regulator that recruits the P-TEFb complex, which includes Cdk9 and cyclin T, to RNA polymerase II (pol II). Together, BRD4 and CDK9 phosphorylate serine 2 (pSer2) of heptad repeats in the C-terminal domain of RPB1, the large subunit of pol II, promote transcriptional elongation. Small-molecule degraders of BRD4 have shown encouraging efficacy in preclinical models for several tumor types but less efficacy in other cancers including small-cell lung cancer (SCLC) and pancreatic cancer. Here, we evaluated CFT-2718, a new BRD4-targeting degrader with enhanced catalytic activity and in vivo properties. In vivo, CFT-2718 has significantly greater efficacy than the CDK9 inhibitor dinaciclib in reducing growth of the LX-36 SCLC patient-derived xenograft (PDX) model and performed comparably to dinaciclib in limiting growth of the PNX-001 pancreatic PDX model. In vitro, CFT-2718 reduced cell viability in four SCLC and two pancreatic cancer models. In SCLC models, this activity significantly exceeded that of dinaciclib; furthermore, CFT-2718 selectively increased the expression of cleaved PARP, an indicator of apoptosis. CFT-2718 caused rapid BRD4 degradation and reduced levels of total and pSer2 RPB1 protein. These and other findings suggest that BRD-mediated transcriptional suppression merits further exploration in the setting of SCLC.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines du cycle cellulaire/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs du poumon/traitement médicamenteux , Tumeurs du pancréas/traitement médicamenteux , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Bibliothèques de petites molécules/pharmacologie , Facteurs de transcription/métabolisme , Animaux , Apoptose , Mouvement cellulaire , Prolifération cellulaire , Femelle , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Souris SCID , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Carcinome pulmonaire à petites cellules/métabolisme , Carcinome pulmonaire à petites cellules/anatomopathologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
2.
SLAS Discov ; 26(4): 547-559, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33780296

RÉSUMÉ

Recent advances in targeted protein degradation have enabled chemical hijacking of the ubiquitin-proteasome system to treat disease. The catalytic rate of cereblon (CRBN)-dependent bifunctional degradation activating compounds (BiDAC), which recruit CRBN to a chosen target protein, resulting in its ubiquitination and proteasomal degradation, is an important parameter to consider during the drug discovery process. In this work, an in vitro system was developed to measure the kinetics of BRD4 bromodomain 1 (BD1) ubiquitination by fitting an essential activator kinetic model to these data. The affinities between BiDACs, BD1, and CRBN in the binary complex, ternary complex, and full ubiquitination complex were characterized. Together, this work provides a new tool for understanding and optimizing the catalytic and thermodynamic properties of BiDACs.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Dosage biologique , Protéines du cycle cellulaire/métabolisme , Oxindoles/pharmacologie , Phtalimides/pharmacologie , Maturation post-traductionnelle des protéines , Facteurs de transcription/métabolisme , Ubiquitin-protein ligases/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines du cycle cellulaire/composition chimique , Protéines du cycle cellulaire/génétique , Système acellulaire/composition chimique , Système acellulaire/métabolisme , Cellules HeLa , Humains , Cinétique , Oxindoles/synthèse chimique , Phtalimides/synthèse chimique , Proteasome endopeptidase complex/effets des médicaments et des substances chimiques , Liaison aux protéines , Domaines protéiques , Protéolyse/effets des médicaments et des substances chimiques , Thermodynamique , Facteurs de transcription/composition chimique , Facteurs de transcription/génétique , Ubiquitin-protein ligases/génétique , Ubiquitination/effets des médicaments et des substances chimiques
3.
Sci Rep ; 8(1): 17239, 2018 11 22.
Article de Anglais | MEDLINE | ID: mdl-30467317

RÉSUMÉ

The development of new treatments for castrate resistant prostate cancer (CRPC) must address such challenges as intrinsic tumor heterogeneity and phenotypic plasticity. Combined PTEN/TP53 alterations represent a major genotype of CRPC (25-30%) and are associated with poor outcomes. Using tumor-derived, castration-resistant Pten/Tp53 null luminal prostate cells for comprehensive, high-throughput, mechanism-based screening, we identified several vulnerabilities among >1900 compounds, including inhibitors of: PI3K/AKT/mTOR, the proteasome, the cell cycle, heat shock proteins, DNA repair, NFκB, MAPK, and epigenetic modifiers. HSP90 inhibitors were one of the most active compound classes in the screen and have clinical potential for use in drug combinations to enhance efficacy and delay the development of resistance. To inform future design of rational drug combinations, we tested ganetespib, a potent second-generation HSP90 inhibitor, as a single agent in multiple CRPC genotypes and phenotypes. Ganetespib decreased growth of endogenous Pten/Tp53 null tumors, confirming therapeutic activity in situ. Fifteen human CRPC LuCaP PDX-derived organoid models were assayed for responses to 110 drugs, and HSP90 inhibitors (ganetespib and onalespib) were among the select group of drugs (<10%) that demonstrated broad activity (>75% of models) at high potency (IC50 <1 µM). Ganetespib inhibits multiple targets, including AR and PI3K pathways, which regulate mutually compensatory growth and survival signals in some forms of CRPC. Combined with castration, ganetespib displayed deeper PDX tumor regressions and delayed castration resistance relative to either monotherapy. In all, comprehensive data from near-patient models presents novel contexts for HSP90 inhibition in multiple CRPC genotypes and phenotypes, expands upon HSP90 inhibitors as simultaneous inhibitors of oncogenic signaling and resistance mechanisms, and suggests utility for combined HSP90/AR inhibition in CRPC.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Antagonistes du récepteur des androgènes/pharmacologie , Animaux , Benzamides/pharmacologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Génotype , Tests de criblage à haut débit/méthodes , Humains , Isoindoles/pharmacologie , Mâle , Souris , Phosphohydrolase PTEN/métabolisme , Phénotype , Phosphatidylinositol 3-kinases/métabolisme , Prostate/effets des médicaments et des substances chimiques , Prostate/métabolisme , Tumeurs prostatiques résistantes à la castration/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Triazoles/pharmacologie , Protéine p53 suppresseur de tumeur/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
4.
Clin Cancer Res ; 24(24): 6396-6407, 2018 12 15.
Article de Anglais | MEDLINE | ID: mdl-30171047

RÉSUMÉ

PURPOSE: STA-1474, prodrug of the heat shock protein 90 inhibitor (HSP90i) ganetespib, previously demonstrated activity in canine preclinical models of cancer; interestingly, prolonged infusions were associated with improved biologic activity. The purpose of this study was to identify the ideal treatment schedule for HSP90i in preclinical models of KIT-driven malignancies and in dogs with spontaneous mast cell tumors (MCT), where KIT is a known driver. EXPERIMENTAL DESIGN: In vitro and murine xenograft experiments and clinical studies in dogs with MCTs were used to define the effects of HSP90i-dosing regimen on client protein downregulation and antitumor activity. RESULTS: Continuous HSP90 inhibition led to durable destabilization of client proteins in vitro; however, transient exposure required >10× drug for comparable effects. In vivo, KIT was rapidly degraded following a single dose of HSP90i but returned to baseline levels within a day. HSP90 levels increased and stabilized 16 hours after HSP90i and were not elevated following a subsequent near-term exposure, providing a functional pool of chaperone to stabilize proteins and a means for greater therapeutic activity upon HSP90i reexposure. HSP90i administered on days 1 and 2 (D1/D2) demonstrated increased biologic activity compared with D1 treatment in KIT or EGFR-driven murine tumor models. In a trial of dogs with MCT, D1/D2 dosing of HSP90i was associated with sustained KIT downregulation, 50% objective response rate and 100% clinical benefit rate compared with D1 and D1/D4 schedules. CONCLUSIONS: These data provide further evidence that prolonged HSP90i exposure improves biologic activity through sustained downregulation of client proteins.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Leucémie à mastocytes/étiologie , Leucémie à mastocytes/métabolisme , Protéines proto-oncogènes c-kit/métabolisme , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Chiens , Protéines du choc thermique HSP90/génétique , Protéines du choc thermique HSP90/métabolisme , Leucémie à mastocytes/traitement médicamenteux , Leucémie à mastocytes/anatomopathologie , Souris , Oncogènes , Protéolyse , Protéines proto-oncogènes c-kit/génétique , Résultat thérapeutique , Triazoles/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
5.
FASEB J ; 32(5): 2735-2746, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29401581

RÉSUMÉ

Autosomal-dominant polycystic kidney disease (ADPKD) is associated with progressive formation of renal cysts, kidney enlargement, hypertension, and typically end-stage renal disease. In ADPKD, inherited mutations disrupt function of the polycystins (encoded by PKD1 and PKD2), thus causing loss of a cyst-repressive signal emanating from the renal cilium. Genetic studies have suggested ciliary maintenance is essential for ADPKD pathogenesis. Heat shock protein 90 (HSP90) clients include multiple proteins linked to ciliary maintenance. We determined that ganetespib, a clinical HSP90 inhibitor, inhibited proteasomal repression of NEK8 and the Aurora-A activator trichoplein, rapidly activating Aurora-A kinase and causing ciliary loss in vitro. Using conditional mouse models for ADPKD, we performed long-term (10 or 50 wk) dosing experiments that demonstrated HSP90 inhibition caused durable in vivo loss of cilia, controlled cystic growth, and ameliorated symptoms induced by loss of Pkd1 or Pkd2. Ganetespib efficacy was not increased by combination with 2-deoxy-d-glucose, a glycolysis inhibitor showing some promise for ADPKD. These studies identify a new biologic activity for HSP90 and support a cilia-based mechanism for cyst repression.-Nikonova, A. S., Deneka, A. Y., Kiseleva, A. A., Korobeynikov, V., Gaponova, A., Serebriiskii, I. G., Kopp, M. C., Hensley, H. H., Seeger-Nukpezah, T. N., Somlo, S., Proia, D. A., Golemis, E. A. Ganetespib limits ciliation and cystogenesis in autosomal-dominant polycystic kidney disease (ADPKD).


Sujet(s)
Polykystose rénale autosomique dominante/traitement médicamenteux , Triazoles/pharmacologie , Animaux , Aurora kinase A/génétique , Aurora kinase A/métabolisme , Cils vibratiles/génétique , Cils vibratiles/métabolisme , Modèles animaux de maladie humaine , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/génétique , Protéines du choc thermique HSP90/métabolisme , Humains , Souris , Souris knockout , Kinases apparentées à NIMA/génétique , Kinases apparentées à NIMA/métabolisme , Polykystose rénale autosomique dominante/génétique , Polykystose rénale autosomique dominante/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Pyruvate dehydrogenase acetyl-transferring kinase
6.
Oncotarget ; 8(3): 4399-4409, 2017 Jan 17.
Article de Anglais | MEDLINE | ID: mdl-27779106

RÉSUMÉ

The lack of effective treatment modalities is a major problem in pancreatic cancer (PCa), a devastating malignancy that is nearly universally driven by the "undruggable" KRAS and TP53 cancer genes. Poor tumor tissue penetration is the major source of resistance in pancreatic cancer where chemotherapy is the mainstay of treatment. In this study we exploited the selective tumor-targeting properties of the heat shock 90 protein inhibitors as the vehicle for drug delivery to pancreatic tumor tissues. STA-12-8666 is a novel esterase-cleavable conjugate of an HSP90i and a topoisomerase I inhibitor, SN-38. STA-12-8666 selectively binds activated HSP90 and releases its cytotoxic payload resulting in drug accumulation in pancreatic cancer cells in vivo. We investigated the preclinical activity of STA-12-8666 in patient derived xenograft and genetic models of pancreatic cancer.Treatment with STA-12-8666 of the KPC mice (knock-in alleles of LSL-KrasG12D, Tp53fl/fl and Pdx1-Cre transgene) at the advanced stages of pancreatic tumors doubled their survival (49 days vs. 74 days, p=0.008). STA-12-8666 also demonstrated dramatically superior activity in comparison to equimolar doses of irinotecan against 5 patient-derived pancreatic adenocarcinoma xenografts with prolonged remissions in some tumors. Analysis of activity of STA-12-8666 against tumor tissues and matched cell lines demonstrated prolonged accumulation and release of cytotoxic payload in the tumor leading to DNA damage response and cell cycle arrest.Our results provide a proof-of-principle validation that HSP90i-based drug conjugates can overcome the notorious treatment resistance by utilizing the inherently high affinity of pancreatic cancer cells to HSP90 antagonists.


Sujet(s)
Antinéoplasiques/administration et posologie , Camptothécine/analogues et dérivés , Carcinome du canal pancréatique/traitement médicamenteux , Tumeurs du pancréas/traitement médicamenteux , Résorcinol/administration et posologie , Animaux , Antinéoplasiques/pharmacologie , Camptothécine/administration et posologie , Camptothécine/pharmacologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Altération de l'ADN , Humains , Souris , Thérapie moléculaire ciblée , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Protéines proto-oncogènes p21(ras)/génétique , Résorcinol/pharmacologie , Protéine p53 suppresseur de tumeur/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Leuk Lymphoma ; 58(4): 923-931, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-27686857

RÉSUMÉ

HSP90 inhibitors have been shown to kill Epstein-Barr virus (EBV)-infected cells by reducing the level of EBV EBNA-1 and/or LMP1. We treated virus-infected cells with ganetespib, an HSP90 inhibitor currently being evaluated in multiple clinical trials for cancer and found that the drug killed EBV-positive B and T cells and reduced the level of both EBV EBNA-1 and LMP1. Treatment of cells with ganetespib also reduced the level of pAkt. Ganetespib delayed the onset of EBV-positive lymphomas and prolonged survival in SCID mice inoculated with one EBV-transformed B-cell line, but not another B-cell line. The former cell line showed lower levels of EBNA-1 after treatment with ganetespib in vitro. Treatment of a patient with T-cell chronic active EBV with ganetespib reduced the percentage of EBV-positive cells in the peripheral blood. These data indicate that HSP90 inhibitors may have a role in the therapy of certain EBV-associated diseases.


Sujet(s)
Lymphocytes B/effets des médicaments et des substances chimiques , Lymphocytes B/virologie , Infections à virus Epstein-Barr/sang , Herpèsvirus humain de type 4/physiologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/virologie , Triazoles/pharmacologie , Animaux , Lignée de cellules transformées , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Modèles animaux de maladie humaine , Infections à virus Epstein-Barr/virologie , Antigènes nucléaires du virus d'Epstein-Barr/génétique , Antigènes nucléaires du virus d'Epstein-Barr/métabolisme , Femelle , Protéines du choc thermique HSC70/génétique , Protéines du choc thermique HSC70/métabolisme , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Humains , Numération des lymphocytes , Souris , Souris SCID
8.
Oncotarget ; 7(40): 65540-65552, 2016 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-27608846

RÉSUMÉ

Long-term survival in patients with metastatic, relapsed, or recurrent Ewing sarcoma and rhabdomyosarcoma is dismal. Irinotecan, a topoisomerase 1 inhibitor, has activity in these sarcomas, but due to poor bioavailability of its active metabolite (SN-38) has had limited clinical efficacy. In this study we have evaluated the efficacy and toxicity of STA-8666, a novel drug conjugate which uses an HSP90 inhibitor to facilitate intracellular, tumor-targeted delivery of the topoisomerase 1 inhibitor SN-38, thus preferentially delivering and concentrating SN-38 within tumor tissue. We present in vivo evidence from mouse xenograft models that STA-8666 results in more persistent inhibition of topoisomerase 1 and prolonged DNA damage compared to irinotecan. This translates into superior antitumor efficacy and survival in multiple aggressive models of both diseases in mouse xenografts, as well as in an irinotecan-resistant model of pediatric osteosarcoma, demonstrated by dramatic tumor shrinkage, durable remission and prolonged complete regressions following short-term treatment, compared to conventional irinotecan. Gene expression analysis performed on xenograft tumors treated with either irinotecan or STA-8666 showed that STA-8666 affected expression of DNA damage and repair genes more robustly than irinotecan. These results suggest that STA-8666 may be a promising new agent for patients with pediatric-type sarcoma.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Camptothécine/analogues et dérivés , Résorcinol/usage thérapeutique , Rhabdomyosarcome/traitement médicamenteux , Sarcome d'Ewing/traitement médicamenteux , Inhibiteurs de la topoisomérase-I/usage thérapeutique , Animaux , Antinéoplasiques/composition chimique , Camptothécine/composition chimique , Camptothécine/usage thérapeutique , Lignée cellulaire tumorale , Enfant , Altération de l'ADN , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Femelle , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Humains , Irinotécan , Souris , Souris knockout , Souris SCID , Résorcinol/composition chimique , Inhibiteurs de la topoisomérase-I/composition chimique , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Cell Rep ; 16(3): 657-71, 2016 07 19.
Article de Anglais | MEDLINE | ID: mdl-27396341

RÉSUMÉ

Anti-Müllerian hormone (AMH) and its type II receptor AMHR2, both previously thought to primarily function in gonadal tissue, were unexpectedly identified as potent regulators of transforming growth factor (TGF-ß)/bone morphogenetic protein (BMP) signaling and epithelial-mesenchymal transition (EMT) in lung cancer. AMH is a TGF-ß/BMP superfamily member, and AMHR2 heterodimerizes with type I receptors (ALK2, ALK3) also used by the type II receptor for BMP (BMPR2). AMH signaling regulates expression of BMPR2, ALK2, and ALK3, supports protein kinase B-nuclear factor κB (AKT-NF-κB) and SMAD survival signaling, and influences BMP-dependent signaling in non-small cell lung cancer (NSCLC). AMH and AMHR2 are selectively expressed in epithelial versus mesenchymal cells, and loss of AMH/AMHR2 induces EMT. Independent induction of EMT reduces expression of AMH and AMHR2. Importantly, EMT associated with depletion of AMH or AMHR2 results in chemoresistance but sensitizes cells to the heat shock protein 90 (HSP90) inhibitor ganetespib. Recognition of this AMH/AMHR2 axis helps to further elucidate TGF-ß/BMP resistance-associated signaling and suggests new strategies for therapeutic targeting of EMT.


Sujet(s)
Hormone antimullérienne/métabolisme , Plasticité cellulaire/physiologie , Résistance aux médicaments antinéoplasiques/physiologie , Cellules épithéliales/métabolisme , Cellules épithéliales/physiologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Animaux , Récepteurs de la protéine morphogénique osseuse de type II/métabolisme , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Transition épithélio-mésenchymateuse/physiologie , Régulation de l'expression des gènes/physiologie , Protéines du choc thermique/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Souris , Souris SCID , Facteur de transcription NF-kappa B/métabolisme , Récepteurs peptidiques/métabolisme , Récepteurs TGF-bêta/métabolisme , Transduction du signal/physiologie , Facteur de croissance transformant bêta/métabolisme
10.
Clin Cancer Res ; 22(23): 5876-5886, 2016 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-27354472

RÉSUMÉ

PURPOSE: HSP90 inhibition is well known to sensitize cancer cells to radiation. However, it is currently unknown whether additional radiosensitization could occur in the more clinically relevant setting of chemoradiation (CRT). We used the potent HSP90 inhibitor ganetespib to determine whether it can enhance CRT effects in NSCLC. EXPERIMENTAL DESIGN: We first performed in vitro experiments in various NSCLC cell lines combining radiation with or without ganetespib. Some of these experiments included clonogenic survival assay, DNA damage repair, and cell-cycle analysis, and reverse-phase protein array. We then determined whether chemotherapy affected ganetespib radiosensitization by adding carboplatin-paclitaxel to some of the in vitro and in vivo xenograft experiments. RESULTS: Ganetespib significantly reduced radiation clonogenic survival in a number of lung cancer cell lines, and attenuated DNA damage repair with irradiation. Radiation caused G2-M arrest that was greatly accentuated by ganetespib. Ganetespib with radiation also dose-dependently upregulated p21 and downregulated pRb levels that were not apparent with either drug or radiation alone. However, when carboplatin-paclitaxel was added, ganetespib was only able to radiosensitize some cell lines but not others. This variable in vitro CRT effect was confirmed in vivo using xenograft models. CONCLUSIONS: Ganetespib was able to potently sensitize a number of NSCLC cell lines to radiation but has variable effects when added to platinum-based doublet CRT. For optimal clinical translation, our data emphasize the importance of preclinical testing of drugs in the context of clinically relevant therapy combinations. Clin Cancer Res; 22(23); 5876-86. ©2016 AACR.


Sujet(s)
Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/radiothérapie , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/radiothérapie , Radiosensibilisants/pharmacologie , Triazoles/pharmacologie , Cellules A549 , Animaux , Carboplatine/pharmacologie , Carcinome pulmonaire non à petites cellules/métabolisme , Lignée cellulaire tumorale , Chimioradiothérapie/méthodes , Femelle , Humains , Tumeurs du poumon/métabolisme , Souris , Souris nude , Paclitaxel/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
11.
Clin Cancer Res ; 22(20): 5120-5129, 2016 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-27267850

RÉSUMÉ

PURPOSE: Small cell lung cancer (SCLC) is a highly aggressive disease representing 12% to 13% of total lung cancers, with median survival of <2 years. No targeted therapies have proven effective in SCLC. Although most patients respond initially to cytotoxic chemotherapies, resistance rapidly emerges, response to second-line agents is limited, and dose-limiting toxicities (DLT) are a major issue. This study performs preclinical evaluation of a new compound, STA-8666, in SCLC. EXPERIMENTAL DESIGN: To avoid DLT for useful cytotoxic agents, the recently developed drug STA-8666 combines a chemical moiety targeting active HSP90 (concentrated in tumors) fused via cleavable linker to SN38, the active metabolite of irinotecan. We compare potency and mechanism of action of STA-8666 and irinotecan in vitro and in vivo RESULTS: In two SCLC xenograft and patient-derived xenograft models, STA-8666 was tolerated without side effects up to 150 mg/kg. At this dose, STA-8666 controlled or eliminated established tumors whether used in a first-line setting or in tumors that had progressed following treatment on standard first- and second-line agents for SCLC. At 50 mg/kg, STA-8666 strongly enhanced the action of carboplatin. Pharmacokinetic profiling confirmed durable STA-8666 exposure in tumors compared with irinotecan. STA-8666 induced a more rapid, robust, and stable induction of cell-cycle arrest, expression of signaling proteins associated with DNA damage and cell-cycle checkpoints, and apoptosis in vitro and in vivo, in comparison with irinotecan. CONCLUSIONS: Together, these results strongly support clinical development of STA-8666 for use in the first- or second-line setting for SCLC. Clin Cancer Res; 22(20); 5120-9. ©2016 AACR.


Sujet(s)
Antinéoplasiques d'origine végétale/usage thérapeutique , Camptothécine/analogues et dérivés , Carboplatine/usage thérapeutique , Systèmes de délivrance de médicaments/méthodes , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Tumeurs du poumon/traitement médicamenteux , Résorcinol/usage thérapeutique , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Animaux , Apoptose/effets des médicaments et des substances chimiques , Camptothécine/usage thérapeutique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Altération de l'ADN/effets des médicaments et des substances chimiques , Synergie des médicaments , Femelle , Humains , Irinotécan , Souris , Souris SCID , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Oncoimmunology ; 5(2): e1075114, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-27057446

RÉSUMÉ

Monoclonal antibodies (mAbs) targeting coinhibitory molecules such as PD-1, PD-L1 and CTLA-4 are increasingly used as targets of therapeutic intervention against cancer. While these targets have led to a critical paradigm shift in treatments for cancer, these approaches are also plagued with limitations owing to cancer immune evasion mechanisms and adverse toxicities associated with continuous treatment. It has been difficult to reproduce and develop interventions to these limitations preclinically due to poor reagent efficacy and reagent xenogenecity not seen in human trials. In this study, we investigated adverse effects of repeated administration of PD-1 and PD-L1 mAbs in the murine 4T1 mammary carcinoma model. We observed rapid and fatal hypersensitivity reactions in tumor bearing mice within 30-60 min after 4-5 administrations of PD-L1 or PD-1 mAb but not CTLA-4 antibody treatment. These events occurred only in mice bearing the highly inflammatory 4T1 tumor and did not occur in mice bearing non-inflammatory tumors. We observed that mortality was associated with systemic accumulation of IgG1 antibodies, antibodies specific to the PD-1 mAb, and accumulation of Gr-1high neutrophils in lungs which have been implicated in the IgG mediated pathway of anaphylaxis. Anti-PD-1 associated toxicities were alleviated when PD-1 blockade was combined with the therapeutic HSP90 inhibitor, ganetespib, which impaired immune responses toward the xenogeneic PD-1 mAb. This study highlights a previously uncharacterized fatal hypersensitivity exacerbated by the PD-1/PD-L1 axis in the broadly used 4T1 tumor model as well as an interesting relationship between this particular class of checkpoint blockade and tumor-dependent immunomodulation.

13.
Horm Cancer ; 7(2): 114-26, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26858237

RÉSUMÉ

Targetable molecular drivers for triple-negative breast cancer (TNBC) have been difficult to identify; therefore, standard treatment remains limited to conventional chemotherapy. Recently, new-generation small-molecule Hsp90 inhibitors (e.g., ganetespib and NVP-AUY922) have demonstrated improved safety and activity profiles over the first-generation ansamycin class. In breast cancer, clinical responses have been observed in a subset of TNBC patients following ganetespib monotherapy; however, the underlying biology of Hsp90 inhibitor treatment and tumor response is not well understood. Glucocorticoid receptor (GR) activity in TNBC is associated with chemotherapy resistance. Here, we find that treatment of TNBC cell lines with ganetespib resulted in GR degradation and decreased GR-mediated gene expression. Ganetespib-associated GR degradation also sensitized TNBC cells to paclitaxel-induced cell death both in vitro and in vivo. The beneficial effect of the Hsp90 inhibitor on paclitaxel-induced cytotoxicity was reduced when GR was depleted in TNBC cells but could be recovered with GR overexpression. These findings suggest that GR-regulated anti-apoptotic and pro-proliferative signaling networks in TNBC are disrupted by Hsp90 inhibitors, thereby sensitizing TNBC to paclitaxel-induced cell death. Thus, GR+ TNBC patients may be a subgroup of breast cancer patients who are most likely to benefit from adding an Hsp90 inhibitor to taxane therapy.


Sujet(s)
Antinéoplasiques/administration et posologie , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Paclitaxel/administration et posologie , Récepteurs aux glucocorticoïdes/métabolisme , Triazoles/administration et posologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Animaux , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Souris , Paclitaxel/usage thérapeutique , Triazoles/pharmacologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Oncotarget ; 6(42): 44306-22, 2015 Dec 29.
Article de Anglais | MEDLINE | ID: mdl-26517240

RÉSUMÉ

Despite the common usage of radiotherapy for the treatment of NSCLC, outcomes for these cancers when treated with ionizing radiation (IR) are still unsatisfactory. A better understanding of the mechanisms underlying resistance to IR is needed to design approaches to eliminate the radioresistant cells and prevent tumor recurrence and metastases. Using multiple fractions of IR we generated radioresistant cells from T2821 and T2851 human lung adenocarcinoma cells. The radioresistant phenotypes present in T2821/R and T2851/R cells include multiple changes in DNA repair genes and proteins expression, upregulation of EMT markers, alterations of cell cycle distribution, upregulation of PI3K/AKT signaling and elevated production of growth factors, cytokines, important for lung cancer progression, such as IL-6, PDGFB and SDF-1 (CXCL12). In addition to being radioresistant these cells were also found to be resistant to cisplatin.HSP90 is a molecular chaperone involved in stabilization and function of multiple client proteins implicated in NSCLC cell survival and radioresistance. We examined the effect of ganetespib, a novel HSP90 inhibitor, on T2821/R and T2851/R cell survival, migration and radioresistance. Our data indicates that ganetespib has cytotoxic activity against parental T2821 and T2851 cells and radioresistant T2821/R and T2851/R lung tumor cells. Ganetespib does not affect proliferation of normal human lung fibroblasts. Combining IR with ganetespib completely abrogates clonogenic survival of radioresistant cells.Our data show that HSP90 inhibition can potentiate the effect of radiotherapy and eliminate radioresistant and cisplatin -resistant residual cells, thus it may aid in reducing NSCLC tumor recurrence after fractionated radiotherapy.


Sujet(s)
Adénocarcinome/radiothérapie , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Tumeurs du poumon/radiothérapie , Radiotolérance/effets des médicaments et des substances chimiques , Radiosensibilisants/pharmacologie , Triazoles/pharmacologie , Adénocarcinome/génétique , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Adénocarcinome pulmonaire , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des radiations , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des radiations , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des radiations , Cisplatine/pharmacologie , Relation dose-effet des médicaments , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des radiations , Protéines du choc thermique HSP90/métabolisme , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Phénotype , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/effets des radiations , Facteurs temps
15.
Mol Cancer Ther ; 14(11): 2422-32, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26271675

RÉSUMÉ

The clinical benefits of chemotherapy are commonly offset by insufficient drug exposures, narrow safety margins, and/or systemic toxicities. Over recent decades, a number of conjugate-based targeting approaches designed to overcome these limitations have been explored. Here, we report on an innovative strategy that utilizes HSP90 inhibitor-drug conjugates (HDC) for directed tumor targeting of chemotherapeutic agents. STA-12-8666 is an HDC that comprises an HSP90 inhibitor fused to SN-38, the active metabolite of irinotecan. Mechanistic analyses in vitro established that high-affinity HSP90 binding conferred by the inhibitor backbone could be exploited for conjugate accumulation within tumor cells. In vivo modeling showed that the HSP90 inhibitor moiety was required for selective retention of STA-12-8666, and this enrichment promoted extended release of active SN-38 within the tumor compartment. Indeed, controlled intratumoral payload release by STA-12-8666 contributed to a broad therapeutic window, sustained biomarker activity, and remarkable degree of efficacy and durability of response in multiple cell line and patient-derived xenograft models. Overall, STA-12-8666 has been developed as a unique HDC agent that employs a distinct mechanism of targeted drug delivery to achieve potent and sustained antitumor effects. These findings identify STA-12-8666 as a promising new candidate for evaluation as novel anticancer therapeutic.


Sujet(s)
Antinéoplasiques/pharmacologie , Camptothécine/analogues et dérivés , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Résorcinol/pharmacologie , Triazoles/pharmacologie , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Technique de Western , Camptothécine/composition chimique , Camptothécine/pharmacocinétique , Camptothécine/pharmacologie , Lignée cellulaire tumorale , Femelle , Protéines du choc thermique HSP90/métabolisme , Humains , Irinotécan , Souris de lignée ICR , Souris SCID , Microscopie de fluorescence , Thérapie moléculaire ciblée/méthodes , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Résorcinol/composition chimique , Résorcinol/pharmacocinétique , Inhibiteurs de la topoisomérase-I/administration et posologie , Inhibiteurs de la topoisomérase-I/pharmacocinétique , Inhibiteurs de la topoisomérase-I/pharmacologie , Résultat thérapeutique , Triazoles/administration et posologie , Triazoles/pharmacocinétique , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
16.
Cancers (Basel) ; 7(2): 876-907, 2015 May 22.
Article de Anglais | MEDLINE | ID: mdl-26010604

RÉSUMÉ

The molecular chaperone HSP90 is involved in stabilization and function of multiple client proteins, many of which represent important oncogenic drivers in NSCLC. Utilization of HSP90 inhibitors as radiosensitizing agents is a promising approach. The antitumor activity of ganetespib, HSP90 inhibitor, was evaluated in human lung adenocarcinoma (AC) cells for its ability to potentiate the effects of IR treatment in both in vitro and in vivo. The cytotoxic effects of ganetespib included; G2/M cell cycle arrest, inhibition of DNA repair, apoptosis induction, and promotion of senescence. All of these antitumor effects were both concentration- and time-dependent. Both pretreatment and post-radiation treatment with ganetespib at low nanomolar concentrations induced radiosensitization in lung AC cells in vitro. Ganetespib may impart radiosensitization through multiple mechanisms: such as down regulation of the PI3K/Akt pathway; diminished DNA repair capacity and promotion of cellular senescence. In vivo, ganetespib reduced growth of T2821 tumor xenografts in mice and sensitized tumors to IR. Tumor irradiation led to dramatic upregulation of ß-catenin expression in tumor tissues, an effect that was mitigated in T2821 xenografts when ganetespib was combined with IR treatments. These data highlight the promise of combining ganetespib with IR therapies in the treatment of AC lung tumors.

17.
Cancer Immunol Res ; 3(6): 583-9, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25948551

RÉSUMÉ

The demonstration that immune checkpoint blockade can meaningfully improve outcomes for cancer patients has revolutionized the field of immuno-oncology. New biologic agents targeting specific checkpoints have shown remarkable durability in terms of patient response and, importantly, exhibit clinical activity across a range of human malignancies, including many that have traditionally proven refractory to other immunotherapies. In this rapidly evolving area, a key consideration relates to the identification of novel combinatorial strategies that exploit existing or investigational cancer therapies in order to optimize patient outcomes and the proportion of individuals able to derive benefit from this approach. In this regard, heat-shock protein 90 (HSP90) represents an important emerging target for cancer therapy because its inactivation results in the simultaneous blockade of multiple signaling pathways and can sensitize tumor cells to other anticancer agents. Within the context of immunology, HSP90 plays a dual regulatory role, with its functional inhibition resulting in both immunosuppressive and immunostimulatory effects. In this Cancer Immunology at the Crossroads overview, the anticancer activity profile of targeted HSP90 inhibitors is discussed along with their paradoxical roles in immunology. Overall, we explore the rationale for combining the modalities of HSP90 inhibition and immune checkpoint blockade in order to augment the antitumor immune response in cancer.


Sujet(s)
Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Thérapie moléculaire ciblée , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Protéines du choc thermique HSP90/métabolisme , Humains , Immunomodulation/effets des médicaments et des substances chimiques , Tumeurs/métabolisme
18.
Target Oncol ; 10(2): 235-45, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25077897

RÉSUMÉ

Small molecule inhibitors of epidermal growth factor receptor (EGFR) tyrosine kinase activity, such as erlotinib and gefitinib, revolutionized therapy for non-small cell lung cancer (NSCLC) patients whose tumors harbor activating EGFR mutations. However, mechanisms to overcome the invariable development of acquired resistance to such agents, as well as realizing their full clinical potential within the context of wild-type EGFR (WT-EGFR) disease, remain to be established. Here, the antitumor efficacy of targeted EGFR tyrosine kinase inhibitors (TKIs) and the HSP90 inhibitor ganetespib, alone and in combination, were evaluated in NSCLC. Ganetespib potentiated the efficacy of erlotinib in TKI-sensitive, mutant EGFR-driven NCI-HCC827 xenograft tumors, with combination treatment causing significant tumor regressions. In erlotinib-resistant NCI-H1975 xenografts, concurrent administration of ganetespib overcame erlotinib resistance to significantly improve tumor growth inhibition. Ganetespib co-treatment also significantly enhanced antitumor responses to afatinib in the same model. In WT-EGFR cell lines, ganetespib potently reduced cell viability. In NCI-H1666 cells, ganetespib-induced loss of client protein expression, perturbation of oncogenic signaling pathways, and induction of apoptosis translated to robust single-agent activity in vivo. Dual ganetespib/erlotinib therapy induced regressions in NCI-H322 xenograft tumors, indicating that the sensitizing properties of ganetespib for erlotinib were conserved within the WT-EGFR setting. Mechanistically, combined ganetespib/erlotinib exposure stabilized EGFR protein levels in an inactive state and completely abrogated extracellular-signal-regulated kinase (ERK) and AKT signaling activity. Thus, selective HSP90 blockade by ganetespib represents a potentially important complementary strategy to targeted TKI inhibition alone for inducing substantial antitumor responses and overcoming resistance, in both the mutant and WT-EGFR settings.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Récepteurs ErbB/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Tumeurs du poumon/traitement médicamenteux , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Triazoles/pharmacologie , Animaux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Résistance aux médicaments antinéoplasiques , Synergie des médicaments , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Femelle , Protéines du choc thermique HSP90/métabolisme , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris SCID , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs temps , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Clin Colorectal Cancer ; 13(4): 207-12, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25444464

RÉSUMÉ

BACKGROUND: Heat shock protein 90 (Hsp90) is a cellular chaperone that is required for the maturation and stability of a variety of proteins that play key roles in colon cancer initiation and progression. The primary objective of the current study was to define the safety and efficacy of ganetespib, a novel, selective small-molecule Hsp90 inhibitor, in patients with refractory metastatic colorectal cancer. PATIENTS AND METHODS: The study was a single-arm, Simon 2-stage, phase II trial for patients with chemotherapy-refractory, metastatic colorectal cancer. Patients received ganetespib 200 mg/m(2) intravenously. Tumor tissue was collected before treatment and 48 hours after treatment for changes in expression of Hsp90 client proteins and other potential pharmacodynamics markers. V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS), v-Raf murine sarcoma viral oncogene homolog B, and phosphatidylinositol-4, 5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) mutational status was also determined. RESULTS: Seventeen patients were treated (median age, 58; range, 44-79 years). No patients demonstrated objective regression of disease. Two patients had stable disease of 6.8 and 5.1 months duration. Serious adverse events that were potentially attributable to ganetespib included diarrhea (12%, n = 2), fatigue (17%, n = 3), and increased aspartate aminotransferase/alanine aminotransferase (12%, n = 2) and alkaline phosphatase (6%, n = 1) levels. Of the 17 evaluable patients, 9 (53%) including patients with stable disease as best response, had KRAS-mutant tumors. CONCLUSION: In this first phase II investigation of an Hsp90 inhibitor in colorectal cancer, ganetespib as a single agent did not demonstrate activity in chemotherapy-refractory metastatic colorectal cancer. However, on the basis of the drug's promising preclinical combination data and the relatively mild toxicity profile, further clinical investigation of this agent in combination with standard cytotoxic agents is planned.


Sujet(s)
Tumeurs colorectales/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Mutation/génétique , Protéines proto-oncogènes/génétique , Thérapie de rattrapage , Triazoles/usage thérapeutique , Protéines G ras/génétique , Adulte , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Tumeurs colorectales/génétique , Tumeurs colorectales/mortalité , Tumeurs colorectales/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Études de suivi , Humains , Techniques immunoenzymatiques , Mâle , Adulte d'âge moyen , Métastase tumorale , Stadification tumorale , Pronostic , Protéines proto-oncogènes p21(ras) , Taux de survie
20.
Proc Natl Acad Sci U S A ; 111(51): 18297-302, 2014 Dec 23.
Article de Anglais | MEDLINE | ID: mdl-25489079

RÉSUMÉ

The efficacy of hormonal therapies for advanced estrogen receptor-positive breast cancers is limited by the nearly inevitable development of acquired resistance. Efforts to block the emergence of resistance have met with limited success, largely because the mechanisms underlying it are so varied and complex. Here, we investigate a new strategy aimed at the very processes by which cancers evolve resistance. From yeast to vertebrates, heat shock protein 90 (HSP90) plays a unique role among molecular chaperones by promoting the evolution of heritable new traits. It does so by regulating the folding of a diverse portfolio of metastable client proteins, many of which mediate adaptive responses that allow organisms to adapt and thrive in the face of diverse challenges, including those posed by drugs. Guided by our previous work in pathogenic fungi, in which very modest HSP90 inhibition impairs resistance to mechanistically diverse antifungals, we examined the effect of similarly modest HSP90 inhibition on the emergence of resistance to antiestrogens in breast cancer models. Even though this degree of inhibition fell below the threshold for proteotoxic activation of the heat-shock response and had no overt anticancer activity on its own, it dramatically impaired the emergence of resistance to hormone antagonists both in cell culture and in mice. Our findings strongly support the clinical testing of combined hormone antagonist-low-level HSP90 inhibitor regimens in the treatment of metastatic estrogen receptor-positive breast cancer. At a broader level, they also provide promising proof of principle for a generalizable strategy to combat the pervasive problem of rapidly emerging resistance to molecularly targeted therapeutics.


Sujet(s)
Antinéoplasiques hormonaux/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Protéines du choc thermique HSP90/physiologie , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Cycle cellulaire , Résistance aux médicaments antinéoplasiques , Femelle , Hétérogreffes , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...