Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Oral Maxillofac Pathol ; 28(1): 134-137, 2024.
Article in English | MEDLINE | ID: mdl-38800424

ABSTRACT

Oncocytic mucoepidermoid carcinoma (OMEC) is an uncommon variant of mucoepidermoid carcinoma. Histopathologically, it is characterised by the predominance of cells with large polygonal morphology and with an abundance of eosinophilic granules. We present a rare case of OMEC manifested as painless palatal swelling in a 25-year-old young male. The overlying mucosa was normal in appearance, with no evidence of ulceration or discharge. Histopathology examination showed the presence of sheets of mucous and intermediate cells along with cystic areas of variable sizes and shapes. On high power magnification, oncocytes were evident, showing abundant granular eosinophilic cytoplasm with central dark round nuclei. Around 75-80% tumour cell population was composed of oncocytic cells. The predominant presence of oncocytes can present diagnostic difficulties to pathologists due to overlapping features with adenoid cystic carcinoma, oncocytoma, acinic cell carcinoma, Warthin's tumour, and other oncocyte tumours. Although the presence of oncocytes is a pathognomonic feature, the role of immunohistochemistry and genetic analysis in diagnosis is discussed in the present paper. Moreover, considering its behaviour as a low-grade MEC, it is prudent to avoid an aggressive treatment strategy and prevent unwarranted morbidity. We recommend prospective studies to better understand the factors that influence the prognosis of OMEC.

3.
Cancer Res ; 82(10): 1858-1869, 2022 05 16.
Article in English | MEDLINE | ID: mdl-35298624

ABSTRACT

Antibody-drug conjugates (ADC) have emerged as one of the pillars of clinical disease management in oncology. The biggest hurdle to widespread development and application of ADCs has been a narrow therapeutic index. Advances in antibody technologies and formats as well as novel linker and payload chemistries have begun to facilitate structural improvements to ADCs. However, the interplay of structural characteristics with physiologic and pharmacologic factors determining therapeutic success has garnered less attention. This review elaborates on the pharmacology of ADCs, the pathophysiology of cancerous tissues, and the reciprocal consequences on ADC properties and functions. While most currently approved ADCs utilize either microtubule inhibition or DNA damage as primary mechanisms of action, we present arguments to expand this repertoire and highlight the need for payload mechanisms that exploit disease-specific vulnerabilities. We promote the idea that the choice of antibody format, targeting antigen, linker properties, and payload of an ADC should be deliberately fit for purpose by taking the pathophysiology of disease and the specific pharmacology of the drug entity into account, thus allowing a higher probability of clinical success.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Antibodies/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Neoplasms/drug therapy
6.
J Neurooncol ; 152(2): 233-243, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33517558

ABSTRACT

PURPOSE: Depatux-m is an antibody drug conjugate (ADC) that targets and inhibits growth of cancer cells overexpressing the epidermal growth factor receptor (EGFR) or the 2-7 deletion mutant (EGFRvIII) in tumor models in vitro and in vivo. Treatment of patients suffering from relapsed/refractory glioblastoma (GBM) with a combination of depatux-m and temozolomide (TMZ) tended to increase overall survival. As a first step to understand the nature of the interaction between the two drugs, we investigated whether the interaction was synergistic, additive or antagonistic. METHODS: The efficacy of ADCs, antibodies, TMZ and radiation was tested in xenograft models of GBM, U-87MG and U-87MG EGFRvIII. Both models express EGFR. U-87MG EGFRvIII was transduced to express EGFRvIII. Changes in tumor volume, biomarkers of cell death and apoptosis after treatment were used to measure efficacy of the various treatments. Synergism of depatux-m and TMZ was verified in three-dimensional cultures of U-87MG and U-87MG EGFRvIII by the method of Chou and Talalay. RESULTS: Combined with TMZ and radiotherapy (RT), depatux-m inhibited xenograft growth of U-87MG and U-87MG EGFRvIII more than either treatment with depatux-m or TMZ + RT. Durability of the response to depatux-m + TMZ + RT or depatux-m + TMZ was more pronounced in U-87MG EGFRvIII than in U-87MG. Efficacy of depatux-m + TMZ was synergistic in U-87MG EGFRvIII and additive in U-87MG. CONCLUSION: Adding depatux-m enhances the efficacy of standard of care therapy in preclinical models of GBM. Durability of response to depatux-m + TMZ in vivo and synergy of the drug-drug interaction correlates with the amount of antigen expressed by the tumor cells.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms , Glioblastoma , Temozolomide/pharmacology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Drug Synergism , ErbB Receptors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 19(10): 2117-2125, 2020 10.
Article in English | MEDLINE | ID: mdl-32847977

ABSTRACT

ABBV-321 (serclutamab talirine), a next-generation EGFR-targeted antibody-drug conjugate (ADC) incorporates a potent pyrrolobenzodiazepine (PBD) dimer toxin conjugated to the EGFR-targeting ABT-806 affinity-matured AM1 antibody. ABBV-321 follows the development of related EGFR-targeted ADCs including depatuxizumab mafodotin (depatux-m, ABT-414), ABT-806 conjugated to monomethyl auristatin F (MMAF), and ABBV-221 (losatuxizumab vedotin), AM1 antibody conjugated to monomethyl auristatin E (MMAE). The distinct tumor selectivity of ABBV-321 differentiates it from many previous highly active antibody PBD conjugates that lack a therapeutic window. Potency of the PBD dimer, combined with increased binding of AM1 to EGFR-positive tumor cells, opens the possibility to target a wide array of tumors beyond those with high levels of EGFR overexpression or amplification, including those insensitive to auristatin-based ADCs. ABBV-321 exhibits potent antitumor activity in cellular and in vivo studies including xenograft cell line and patient-derived xenograft glioblastoma, colorectal, lung, head and neck, and malignant mesothelioma tumor models that are less sensitive to depatux-m or ABBV-221. Combination studies with ABBV-321 and depatux-m suggest a promising treatment option permitting suboptimal, and potentially better tolerated, doses of both ADCs while providing improved potency. Collectively, these data suggest that ABBV-321 may offer an extended breadth of efficacy relative to other EGFR ADCs while extending utility to multiple EGFR-expressing tumor indications. Despite its highly potent PBD dimer payload, the tumor selectivity of ABBV-321, coupled with its pharmacology, toxicology, and pharmacokinetic profiles, support continuation of ongoing phase I clinical trials in patients with advanced EGFR-expressing malignancies.


Subject(s)
ErbB Receptors/metabolism , Immunoconjugates/therapeutic use , Animals , Cell Line, Tumor , Female , Humans , Immunoconjugates/pharmacology , Mice , Mice, Nude
8.
J Maxillofac Oral Surg ; 19(3): 387-393, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32801533

ABSTRACT

BACKGROUND: There are scarcely any reported cases of unilateral dislocation in the literature. Hence, its etiology, a possible mechanism of injury, clinical features, and effective management strategies are yet to be described. CASE: A 27-year-old male patient presented with the dilemma of unilateral dislocation of left TMJ. This was addressed by the use of a novel technique in dislocation management. Here, the author also proposes a modified classification system for the TMJ dislocation. CONCLUSION: "Wagh-Kokane's" technique of manual reduction should be encouraged in complex cases of dislocation.

9.
Indian J Otolaryngol Head Neck Surg ; 71(Suppl 3): 2087-2090, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31763299

ABSTRACT

Ossifying fibroma is a benign fibro-osseous lesion found exclusively in jaws. It has a predilection for premolar-molar region in the mandible. The occurrence of OF as solitary lesions with no underlying disease is common in jaws. However its co-existence in patients with neurofibromatosis type 1 (NF1) has not been described in jaws. NF1, also known as von Recklinghausen's disease or peripheral neurofibromatosis, is an autosomal dominant multisystem disorder that approximately affects 1 in 2500-3000 births. The common manifestations of this disease include Café-au-lait macules, skinfold freckling, cutaneous neurofibromas, blue-red and pseudoatrophic macules on skin, plexiform neurofibroma, scoliosis, optic glioma. So far only one case of ossifying fibroma (OF) in such patients has been reported in the skull but not in the maxillofacial region. We report a case of OF of the maxilla in a 45 year old female suffering from NF1. To the best of our knowledge this is the first case report where OF occurred in the maxilla in patient with NF1.

10.
Indian J Otolaryngol Head Neck Surg ; 71(Suppl 1): 748-751, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31742056

ABSTRACT

Osteosarcoma is a mesenchymal malignant tumour of long bones; it is the second most common malignancy of bone after multiple myeloma but it rarely affects jaw, accounting only for 4-8% of all osteosarcomas (Baumhoer et al. in Oral Oncol 50(2):147-153, 2014). The rare occurrence of tumour makes it challenging to diagnose either by radiographic or histopathologic examination. Here we presented a rare case of chondroblastic variant of osteosarcoma of body of mandible in a 54 year old female.

11.
Ann Maxillofac Surg ; 8(1): 28-34, 2018.
Article in English | MEDLINE | ID: mdl-29963421

ABSTRACT

CONTEXT AND AIM: In today's world of advanced dentistry, there are various aspects of restorative, esthetic, and surgical processes. Healing of an extraction socket comprises of bone as well as soft-tissue remodeling with maximum dimensional changes occurring during the first 3 months. Platelet-rich fibrin (PRF) was first developed in France as a therapeutic alternative to platelet-rich plasma to overcome many of its limitations. The present study was planned to evaluate and compare wound healing and bone regeneration in extraction sockets with and without PRF. MATERIALS AND METHODS: The present study was carried out on 30 patients selected from the outpatient department over a period of 2½ years starting from May 2013 undergoing extraction of maxillary or mandibular teeth simultaneously to conduct a split-mouth study. The research protocol was approved by the Institutional Ethics Committee governing the use of human subjects in clinical experimentation. STATISTICAL ANALYSIS USED: Descriptive and analytical statistics were calculated using Statistical Package for Social Sciences version 19. Chi-square test was used to assess wound healing score in the two groups while paired t-test was used to compare the bone density in the socket and periapical regions at different time intervals, and unpaired t-test was used for the intergroup comparisons. P < 0.05 was considered to be significant while P < 0.001 was considered highly significant. RESULTS: Patients in PRF group had better healing index when compared to without PRF group. Use of PRF showed a comparable increase in bone density too. CONCLUSION: An appreciable wound healing and bone regeneration was seen in the experimental group when compared to the control sites where no PRF was used substantiating the use of PRF as an inexpensive autologous material for socket preservation and future rehabilitation. The present study, also, showed that minimal operator expertise was required to conduct the procedure of PRF preparation and grafting when compared to bone harvesting from distant sites. The shorter duration between extractions and further rehabilitation obviates the need for a second procedure.

12.
Mol Cancer Ther ; 17(4): 795-805, 2018 04.
Article in English | MEDLINE | ID: mdl-29483208

ABSTRACT

Depatuxizumab mafodotin (depatux-m, ABT-414) is a tumor-selective antibody drug conjugate (ADC) comprised of the anti-EGFR antibody ABT-806 and the monomethyl auristatin F (MMAF) warhead. Depatux-m has demonstrated promising clinical activity in glioblastoma multiforme (GBM) patients and is currently being evaluated in clinical trials in first-line and recurrent GBM disease settings. Depatux-m responses have been restricted to patients with amplified EGFR, highlighting the need for therapies with activity against tumors with nonamplified EGFR overexpression. In addition, depatux-m dosing has been limited by corneal side effects common to MMAF conjugates. We hypothesized that a monomethyl auristatin E (MMAE) ADC utilizing an EGFR-targeting antibody with increased affinity may have broader utility against tumors with more modest EGFR overexpression while mitigating the risk of corneal side effects. We describe here preclinical characterization of ABBV-221, an EGFR-targeting ADC comprised of an affinity-matured ABT-806 conjugated to MMAE. ABBV-221 binds to a similar EGFR epitope as depatux-m and retains tumor selectivity with increased binding to EGFR-positive tumor cells and greater in vitro potency. ABBV-221 displays increased tumor uptake and antitumor activity against wild-type EGFR-positive xenografts with a greatly reduced incidence of corneal side effects relative to depatux-m. ABBV-221 has similar activity as depatux-m against an EGFR-amplified GBM patient derived xenograft (PDX) model and is highly effective alone and in combination with standard-of-care temozolomide in an EGFRvIII-positive GBM xenograft model. Based on these results, ABBV-221 has advanced to a phase I clinical trial in patients with advanced solid tumors associated with elevated levels of EGFR. Mol Cancer Ther; 17(4); 795-805. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Glioblastoma/drug therapy , Immunoconjugates/pharmacology , Oligopeptides/chemistry , Animals , Antibodies, Monoclonal, Humanized/chemistry , Apoptosis , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunoconjugates/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Neoplasia ; 19(9): 695-706, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28787674

ABSTRACT

Improving the congruity of preclinical models with cancer as it is manifested in humans is a potential way to mitigate the high attrition rate of new cancer therapies in the clinic. In this regard, three-dimensional (3D) tumor cultures in vitro have recently regained interest as they have been acclaimed to have higher similarity to tumors in vivo than to cells grown in monolayers (2D). To identify cancer functions that are active in 3D rather than in 2D cultures, we compared the transcriptional profiles (TPs) of two non-small cell lung carcinoma cell lines, NCI-H1650 and EBC-1 grown in both conditions to the TP of xenografted tumors. Because confluence, diameter or volume can hypothetically alter TPs, we made intra- and inter-culture comparisons using samples with defined dimensions. As projected by Ingenuity Pathway Analysis (IPA), a limited number of signal transduction pathways operational in vivo were better represented by 3D than by 2D cultures in vitro. Growth of 2D and 3D cultures as well as xenografts induced major changes in the TPs of these 3 modes of culturing. Alterations of transcriptional network activation that were predicted to evolve similarly during progression of 3D cultures and xenografts involved the following functions: hypoxia, proliferation, cell cycle progression, angiogenesis, cell adhesion, and interleukin activation. Direct comparison of TPs of 3D cultures and xenografts to monolayer cultures yielded up-regulation of networks involved in hypoxia, TGF and Wnt signaling as well as regulation of epithelial mesenchymal transition. Differences in TP of 2D and 3D cancer cell cultures are subject to progression of the cultures. The emulation of the predicted cell functions in vivo is therefore not only determined by the type of culture in vitro but also by the confluence or diameter of the 2D or 3D cultures, respectively. Consequently, the successful implementation of 3D models will require phenotypic characterization to verify the relevance of applying these models for drug development.


Subject(s)
Gene Expression Regulation, Neoplastic , Transcriptome , Animals , Cell Culture Techniques , Cell Line, Tumor , Cluster Analysis , Disease Models, Animal , Female , Gene Expression Profiling , Heterografts , Humans , Mice , Spheroids, Cellular
15.
Pharmacology ; 100(5-6): 229-242, 2017.
Article in English | MEDLINE | ID: mdl-28743107

ABSTRACT

ABT-700 is a therapeutic antibody against the hepatocyte growth factor receptor (MET). At doses or regimens that lead to exposures exceeding optimum in vivo, the efficacy of ABT-700 is unexpectedly reduced. We hypothesized that this reduction in efficacy was due to a "prozone-like" effect in vivo. A prozone-like effect, which is a reduction in efficacy beyond optimum exposure, is caused due a mechanism similar to the generation of false negative flocculation tests by excessive antibody titres. In vitro, we demonstrate that at higher ABT-700 concentrations, this "prozone-like" effect is mediated by a progressive conversion from bivalent to ineffective monovalent binding of the antibody. In vivo, the efficacy of ABT-700 is dependent on an optimum range of exposure as well. Our data suggest that the "prozone-like" effect is operative and independent of target expression. ABT-700 dose, regimen, exposure, and tumor burden are interdependent variables influencing the "prozone-like" effect and mediating and in vivo efficacy. By optimization of dosage and regimen we demonstrate that the "prozone-like" effect can be alleviated and ABT-700 efficacy at varying tumor loads can be further extended in combination with cisplatin. Our results suggest that optimization of exposure taking tumor burden into account may alleviate "prozone-like" effects without compromising efficacy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Cell Line , Cisplatin/administration & dosage , Humans , Mice , Mice, Nude , Mice, SCID
16.
Clin Cancer Res ; 23(4): 992-1000, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27573171

ABSTRACT

Purpose: Despite the importance of the MET oncogene in many malignancies, clinical strategies targeting c-Met have benefitted only small subsets of patients with tumors driven by signaling through the c-Met pathway, thereby necessitating selection of patients with MET amplification and/or c-Met activation most likely to respond. An ADC targeting c-Met could overcome these limitations with potential as a broad-acting therapeutic.Experimental Design: ADC ABBV-399 was generated with the c-Met-targeting antibody, ABT-700. Antitumor activity was evaluated in cancer cells with overexpressed c-Met or amplified MET and in xenografts including patient-derived xenograft (PDX) models and those refractory to other c-Met inhibitors. The correlation between c-Met expression and sensitivity to ABBV-399 in tumor and normal cell lines was assessed to evaluate the risk of on-target toxicity.Results: A threshold level of c-Met expressed by sensitive tumor but not normal cells is required for significant ABBV-399-mediated killing of tumor cells. Activity extends to c-Met or amplified MET cell line and PDX models where significant tumor growth inhibition and regressions are observed. ABBV-399 inhibits growth of xenograft tumors refractory to other c-Met inhibitors and provides significant therapeutic benefit in combination with standard-of-care chemotherapy.Conclusions: ABBV-399 represents a novel therapeutic strategy to deliver a potent cytotoxin to c-Met-overexpressing tumor cells enabling cell killing regardless of reliance on MET signaling. ABBV-399 has progressed to a phase I study where it has been well tolerated and has produced objective responses in c-Met-expressing non-small cell lung cancer (NSCLC) patients. Clin Cancer Res; 23(4); 992-1000. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-met/genetics , Animals , Antibodies, Monoclonal/adverse effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Neoplasms/immunology , Neoplasms/pathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
17.
BMC Cancer ; 16: 105, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26879245

ABSTRACT

BACKGROUND: c-Met is the receptor tyrosine kinase for hepatocyte growth factor (HGF) encoded by the MET proto-oncogene. Aberrant activation of c-Met resulting from MET amplification and c-Met overexpression is associated with poor clinical outcome in multiple malignancies underscoring the importance of c-Met signaling in cancer progression. Several c-Met inhibitors have advanced to the clinic; however, the development of inhibitory c-Met-directed therapeutic antibodies has been hampered by inherent agonistic activity. METHOD: We generated and tested a bivalent anti-c-Met monoclonal antibody ABT-700 in vitro for binding potency and antagonistic activity and in vivo for antitumor efficacy in human tumor xenografts. Human cancer cell lines and gastric cancer tissue microarrays were examined for MET amplification by fluorescence in situ hybridization (FISH). RESULTS: ABT-700 exhibits a distinctive ability to block both HGF-independent constitutive c-Met signaling and HGF-dependent activation of c-Met. Cancer cells addicted to the constitutively activated c-Met signaling driven by MET amplification undergo apoptosis upon exposure to ABT-700. ABT-700 induces tumor regression and tumor growth delay in preclinical tumor models of gastric and lung cancers harboring amplified MET. ABT-700 in combination with chemotherapeutics also shows additive antitumor effect. Amplification of MET in human cancer tissues can be identified by FISH. CONCLUSIONS: The preclinical attributes of ABT-700 in blocking c-Met signaling, inducing apoptosis and suppressing tumor growth in cancers with amplified MET provide rationale for examining its potential clinical utility for the treatment of cancers harboring MET amplification.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-met/drug effects , Proto-Oncogene Proteins c-met/genetics , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Gene Amplification , Humans , Male , Mice , Mice, SCID , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Xenograft Model Antitumor Assays
18.
Mol Cancer Ther ; 15(4): 661-9, 2016 04.
Article in English | MEDLINE | ID: mdl-26846818

ABSTRACT

Targeting tumor-overexpressed EGFR with an antibody-drug conjugate (ADC) is an attractive therapeutic strategy; however, normal tissue expression represents a significant toxicity risk. The anti-EGFR antibody ABT-806 targets a unique tumor-specific epitope and exhibits minimal reactivity to EGFR in normal tissue, suggesting its suitability for the development of an ADC. We describe the binding properties and preclinical activity of ABT-414, an ABT-806 monomethyl auristatin F conjugate. In vitro, ABT-414 selectively kills tumor cells overexpressing wild-type or mutant forms of EGFR. ABT-414 inhibits the growth of xenograft tumors with high EGFR expression and causes complete regressions and cures in the most sensitive models. Tumor growth inhibition is also observed in tumor models with EGFR mutations, including activating mutations and those with the exon 2-7 deletion [EGFR variant III (EGFRvIII)], commonly found in glioblastoma multiforme. ABT-414 exhibits potent cytotoxicity against glioblastoma multiforme patient-derived xenograft models expressing either wild-type EGFR or EGFRvIII, with sustained regressions and cures observed at clinically relevant doses. ABT-414 also combines with standard-of-care treatment of radiation and temozolomide, providing significant therapeutic benefit in a glioblastoma multiforme xenograft model. On the basis of these results, ABT-414 has advanced to phase I/II clinical trials, and objective responses have been observed in patients with both amplified wild-type and EGFRvIII-expressing tumors. Mol Cancer Ther; 15(4); 661-9. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Epitopes , ErbB Receptors/antagonists & inhibitors , Immunoconjugates/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Antibody Affinity , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , Disease Models, Animal , Epitopes/immunology , ErbB Receptors/genetics , ErbB Receptors/immunology , ErbB Receptors/metabolism , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Molecular Targeted Therapy , Mutation , Protein Binding , Xenograft Model Antitumor Assays
19.
Sci Transl Med ; 7(279): 279ra40, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25787766

ABSTRACT

The BCL-2/BCL-XL/BCL-W inhibitor ABT-263 (navitoclax) has shown promising clinical activity in lymphoid malignancies such as chronic lymphocytic leukemia. However, its efficacy in these settings is limited by thrombocytopenia caused by BCL-XL inhibition. This prompted the generation of the BCL-2-selective inhibitor venetoclax (ABT-199/GDC-0199), which demonstrates robust activity in these cancers but spares platelets. Navitoclax has also been shown to enhance the efficacy of docetaxel in preclinical models of solid tumors, but clinical use of this combination has been limited by neutropenia. We used venetoclax and the BCL-XL-selective inhibitors A-1155463 and A-1331852 to assess the relative contributions of inhibiting BCL-2 or BCL-XL to the efficacy and toxicity of the navitoclax-docetaxel combination. Selective BCL-2 inhibition suppressed granulopoiesis in vitro and in vivo, potentially accounting for the exacerbated neutropenia observed when navitoclax was combined with docetaxel clinically. By contrast, selectively inhibiting BCL-XL did not suppress granulopoiesis but was highly efficacious in combination with docetaxel when tested against a range of solid tumors. Therefore, BCL-XL-selective inhibitors have the potential to enhance the efficacy of docetaxel in solid tumors and avoid the exacerbation of neutropenia observed with navitoclax. These studies demonstrate the translational utility of this toolkit of selective BCL-2 family inhibitors and highlight their potential as improved cancer therapeutics.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Administration, Oral , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Benzothiazoles/chemistry , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Line, Tumor , Cell Survival , Docetaxel , Gene Expression Profiling , Granulocytes/metabolism , Humans , Isoquinolines/chemistry , Kinetics , Mice , Neoplasm Transplantation , Neoplasms/metabolism , Neutropenia/chemically induced , Neutrophils/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/therapeutic use , Taxoids/adverse effects , Thrombocytopenia/chemically induced , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/metabolism
20.
Article in English | MEDLINE | ID: mdl-26764794

ABSTRACT

In this paper, we show how to use canonical perturbation theory for dissipative dynamical systems capable of showing limit-cycle oscillations. Thus, our work surmounts the hitherto perceived barrier for canonical perturbation theory that it can be applied only to a class of conservative systems, viz., Hamiltonian systems. In the process, we also find Hamiltonian structure for an important subset of Liénard system-a paradigmatic system for modeling isolated and asymptotic oscillatory state. We discuss the possibility of extending our method to encompass an even wider range of nonconservative systems.

SELECTION OF CITATIONS
SEARCH DETAIL
...