Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Clin Invest ; 133(22)2023 11 15.
Article in English | MEDLINE | ID: mdl-37725435

ABSTRACT

Antibody-drug conjugates (ADCs) are a promising targeted cancer therapy; however, patient selection based solely on target antigen expression without consideration for cytotoxic payload vulnerabilities has plateaued clinical benefits. Biomarkers to capture patients who might benefit from specific ADCs have not been systematically determined for any cancer. We present a comprehensive therapeutic and biomarker analysis of a B7H3-ADC with pyrrolobenzodiazepine(PBD) payload in 26 treatment-resistant, metastatic prostate cancer (mPC) models. B7H3 is a tumor-specific surface protein widely expressed in mPC, and PBD is a DNA cross-linking agent. B7H3 expression was necessary but not sufficient for B7H3-PBD-ADC responsiveness. RB1 deficiency and/or replication stress, characteristics of poor prognosis, and conferred sensitivity were associated with complete tumor regression in both neuroendocrine (NEPC) and androgen receptor positive (ARPC) prostate cancer models, even with low B7H3 levels. Non-ARPC models, which are currently lacking efficacious treatment, demonstrated the highest replication stress and were most sensitive to treatment. In RB1 WT ARPC tumors, SLFN11 expression or select DNA repair mutations in SLFN11 nonexpressors governed response. Importantly, WT TP53 predicted nonresponsiveness (7 of 8 models). Overall, biomarker-focused selection of models led to high efficacy of in vivo treatment. These data enable a paradigm shift to biomarker-driven trial designs for maximizing clinical benefit of ADC therapies.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Prostatic Neoplasms , Male , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Biomarkers, Tumor/genetics , Cell Line, Tumor , Xenograft Model Antitumor Assays , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Nuclear Proteins
2.
Mol Cell Proteomics ; 22(9): 100626, 2023 09.
Article in English | MEDLINE | ID: mdl-37517589

ABSTRACT

The National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium (CPTAC) provides unique opportunities for cancer target discovery using protein expression. Proteomics data from CPTAC tumor types have been primarily generated using a multiplex tandem mass tag (TMT) approach, which is designed to provide protein quantification relative to reference samples. However, relative protein expression data are suboptimal for prioritization of targets within a tissue type, which requires additional reprocessing of the original proteomics data to derive absolute quantitation estimation. We evaluated the feasibility of using differential protein analysis coupled with intensity-based absolute quantification (iBAQ) to identify tumor-enriched and highly expressed cell surface antigens, employing tandem mass tag (TMT) proteomics data from CPTAC. Absolute quantification derived from TMT proteomics data was highly correlated with that of label-free proteomics data from the CPTAC colon adenocarcinoma cohort, which contains proteomics data measured by both approaches. We validated the TMT-iBAQ approach by comparing the iBAQ value to the receptor density value of HER2 and TROP2 measured by flow cytometry in about 30 selected breast and lung cancer cell lines from the Cancer Cell Line Encyclopedia. Collections of these tumor-enriched and highly expressed cell surface antigens could serve as a valuable resource for the development of cancer therapeutics, including antibody-drug conjugates and immunotherapeutic agents.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Humans , Proteomics , Colonic Neoplasms/therapy , Cell Line
3.
Mol Cancer Ther ; 19(8): 1649-1659, 2020 08.
Article in English | MEDLINE | ID: mdl-32404408

ABSTRACT

Multiple myeloma is a hematologic cancer that disrupts normal bone marrow function and has multiple lines of therapeutic options, but is incurable as patients ultimately relapse. We developed a novel antibody-drug conjugate (ADC) targeting CS-1, a protein that is highly expressed on multiple myeloma tumor cells. The anti-CS-1 mAb specifically bound to cells expressing CS-1 and, when conjugated to a cytotoxic pyrrolobenzodiazepine payload, reduced the viability of multiple myeloma cell lines in vitro In mouse models of multiple myeloma, a single administration of the CS-1 ADC caused durable regressions in disseminated models and complete regression in a subcutaneous model. In an exploratory study in cynomolgus monkeys, the CS-1 ADC demonstrated a half-life of 3 to 6 days; however, no highest nonseverely toxic dose was achieved, as bone marrow toxicity was dose limiting. Bone marrow from dosed monkeys showed reductions in progenitor cells as compared with normal marrow. In vitro cell killing assays demonstrated that the CS-1 ADC substantially reduced the number of progenitor cells in healthy bone marrow, leading us to identify previously unreported CS-1 expression on a small population of progenitor cells in the myeloid-erythroid lineage. This finding suggests that bone marrow toxicity is the result of both on-target and off-target killing by the ADC.


Subject(s)
Antibodies, Monoclonal/chemistry , Antineoplastic Agents/pharmacology , Benzodiazepines/chemistry , Immunoconjugates/pharmacology , Membrane Proteins/antagonists & inhibitors , Microfilament Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Pyrroles/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cell Proliferation , Drug Evaluation, Preclinical , Female , Humans , Immunoconjugates/chemistry , Macaca fascicularis , Membrane Proteins/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Microfilament Proteins/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancer Res ; 80(4): 771-783, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31843981

ABSTRACT

Integrin ß4 (ITGB4) has been shown to play an important role in the regulation of cancer stem cells (CSC). Immune targeting of ITGB4 represents a novel approach to target this cell population, with potential clinical benefit. We developed two immunologic strategies to target ITGB4: ITGB4 protein-pulsed dendritic cells (ITGB4-DC) for vaccination and adoptive transfer of anti-CD3/anti-ITGB4 bispecific antibody (ITGB4 BiAb)-armed tumor-draining lymph node T cells. Two immunocompetent mouse models were utilized to assess the efficacy of these immunotherapies in targeting both CSCs and bulk tumor populations: 4T1 mammary tumors and SCC7 head and neck squamous carcinoma cell line. Immunologic targeting of ITGB4 utilizing either ITGB4-DC or ITGB4 BiAb-T cells significantly inhibited local tumor growth and metastases in both the 4T1 and SCC7 tumor models. Furthermore, the efficacy of both of these ITGB4-targeted immunotherapies was significantly enhanced by the addition of anti-PD-L1. Both ITGB4-targeted immunotherapies induced endogenous T-cell cytotoxicity directed at CSCs as well as non-CSCs, which expressed ITGB4, and immune plasma-mediated killing of CSCs. As a result, ITGB4-targeted immunotherapy reduced not only the number of ITGB4high CSCs in residual 4T1 and SCC7 tumors but also their tumor-initiating capacity in secondary mouse implants. In addition, treated mice demonstrated no apparent toxicity. The specificity of these treatments was demonstrated by the lack of effects observed using ITGB4 knockout 4T1 or ITGB4-negative CT26 colon carcinoma cells. Because ITGB4 is expressed by CSCs across a variety of tumor types, these results support immunologic targeting of ITGB4 as a promising therapeutic strategy.Significance: This study identifies a novel mechanism of resistance to anti-PD-1/PD-L1 immunotherapy mediated by HPV E5, which can be exploited using the HPV E5 inhibitor rimantadine to improve outcomes for head and neck cancer patients.


Subject(s)
Cancer Vaccines/administration & dosage , Immunotherapy, Adoptive/methods , Integrin beta4/metabolism , Neoplasms/therapy , Neoplastic Stem Cells/immunology , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD3 Complex/antagonists & inhibitors , CD3 Complex/immunology , Cancer Vaccines/immunology , Carcinogenesis/immunology , Cell Line, Tumor/transplantation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Gene Knockout Techniques , Humans , Integrin beta4/genetics , Integrin beta4/immunology , Lymph Nodes/cytology , Mice , Molecular Targeted Therapy/methods , Neoplasm Metastasis/immunology , Neoplasm Metastasis/prevention & control , Neoplasms/immunology , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
7.
J Clin Invest ; 128(2): 805-815, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29337305

ABSTRACT

Programmed death-1 receptor (PD-L1, B7-H1) and programmed cell death protein 1 (PD-1) pathway blockade is a promising therapy for treating cancer. However, the mechanistic contribution of host and tumor PD-L1 and PD-1 signaling to the therapeutic efficacy of PD-L1 and PD-1 blockade remains elusive. Here, we evaluated 3 tumor-bearing mouse models that differ in their sensitivity to PD-L1 blockade and demonstrated a loss of therapeutic efficacy of PD-L1 blockade in immunodeficient mice and in PD-L1- and PD-1-deficient mice. In contrast, neither knockout nor overexpression of PD-L1 in tumor cells had an effect on PD-L1 blockade efficacy. Human and murine studies showed high levels of functional PD-L1 expression in dendritic cells and macrophages in the tumor microenvironments and draining lymph nodes. Additionally, expression of PD-L1 on dendritic cells and macrophages in ovarian cancer and melanoma patients correlated with the efficacy of treatment with either anti-PD-1 alone or in combination with anti-CTLA-4. Thus, PD-L1-expressing dendritic cells and macrophages may mechanistically shape and therapeutically predict clinical efficacy of PD-L1/PD-1 blockade.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cytokines/metabolism , Dendrites/metabolism , Dendritic Cells/metabolism , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Lymph Nodes/pathology , Macrophages/metabolism , Male , Melanoma/drug therapy , Melanoma/pathology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, SCID , Ovarian Neoplasms/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Time Factors , Tumor Microenvironment
8.
Oncotarget ; 8(39): 65090-65099, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29029414

ABSTRACT

ADAM17 (a disintegrin and metalloproteinase 17)/TACE (TNFα converting enzyme) has emerged as a potential therapeutic target in colorectal cancer (CRC) and other cancers, due in part to its role in regulating various tumor cell surface proteins and growth factors and cytokines in the tumor microenvironment. The emergence of MEDI3622, a highly potent and specific antibody-based ADAM17 inhibitor, has allowed testing of the concept that targeting ADAM17 may be an important new therapeutic approach for CRC patients. We demonstrate that MEDI3622 is highly efficacious on tumor growth in multiple human CRC PDX models, resulting in improved survival of animals bearing tumor xenografts. MEDI3622 was further found to impact Notch pathway activity and tumor-initiating cells. The promising preclinical activity seen here supports further clinical investigation of this treatment approach to improve therapeutic outcome for patients diagnosed with metastatic CRC, including patients with KRAS-mutant tumors for whom other therapeutic options are currently limited.

9.
Mol Cancer Ther ; 16(8): 1576-1587, 2017 08.
Article in English | MEDLINE | ID: mdl-28522587

ABSTRACT

Antibody-drug conjugates (ADC) are used to selectively deliver cytotoxic agents to tumors and have the potential for increased clinical benefit to cancer patients. 5T4 is an oncofetal antigen overexpressed on the cell surface in many carcinomas on both bulk tumor cells as well as cancer stem cells (CSC), has very limited normal tissue expression, and can internalize when bound by an antibody. An anti-5T4 antibody was identified and optimized for efficient binding and internalization in a target-specific manner, and engineered cysteines were incorporated into the molecule for site-specific conjugation. ADCs targeting 5T4 were constructed by site-specifically conjugating the antibody with payloads that possess different mechanisms of action, either a DNA cross-linking pyrrolobenzodiazepine (PBD) dimer or a microtubule-destabilizing tubulysin, so that each ADC had a drug:antibody ratio of 2. The resulting ADCs demonstrated significant target-dependent activity in vitro and in vivo; however, the ADC conjugated with a PBD payload (5T4-PBD) elicited more durable antitumor responses in vivo than the tubulysin conjugate in xenograft models. Likewise, the 5T4-PBD more potently inhibited the growth of 5T4-positive CSCs in vivo, which likely contributed to its superior antitumor activity. Given that the 5T4-PBD possessed both potent antitumor activity as well as anti-CSC activity, and thus could potentially target bulk tumor cells and CSCs in target-positive indications, it was further evaluated in non-GLP rat toxicology studies that demonstrated excellent in vivo stability with an acceptable safety profile. Taken together, these preclinical data support further development of 5T4-PBD, also known as MEDI0641, against 5T4+ cancer indications. Mol Cancer Ther; 16(8); 1576-87. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Benzodiazepines/therapeutic use , Immunoconjugates/therapeutic use , Pyrroles/therapeutic use , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Benzodiazepines/adverse effects , Benzodiazepines/pharmacology , Cell Line, Tumor , Humans , Immunoconjugates/adverse effects , Immunoconjugates/pharmacology , Male , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pyrroles/adverse effects , Pyrroles/pharmacology , Rats, Sprague-Dawley , Tubulin Modulators/adverse effects , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 23(10): 2516-2527, 2017 May 15.
Article in English | MEDLINE | ID: mdl-27780858

ABSTRACT

Purpose: Locoregional recurrence is a frequent treatment outcome for patients with advanced head and neck squamous cell carcinoma (HNSCC). Emerging evidence suggests that tumor recurrence is mediated by a small subpopulation of uniquely tumorigenic cells, that is, cancer stem cells (CSC), that are resistant to conventional chemotherapy, endowed with self-renewal and multipotency.Experimental Design: Here, we evaluated the efficacy of MEDI0641, a novel antibody-drug conjugate targeted to 5T4 and carrying a DNA-damaging "payload" (pyrrolobenzodiazepine) in preclinical models of HNSCC.Results: Analysis of a tissue microarray containing 77 HNSCC with follow-up of up to 12 years revealed that patients with 5T4high tumors displayed lower overall survival than those with 5T4low tumors (P = 0.038). 5T4 is more highly expressed in head and neck CSC (ALDHhighCD44high) than in control cells (non-CSC). Treatment with MEDI0641 caused a significant reduction in the CSC fraction in HNSCC cells (UM-SCC-11B, UM-SCC-22B) in vitro Notably, a single intravenous dose of 1 mg/kg MEDI0641 caused long-lasting tumor regression in three patient-derived xenograft (PDX) models of HNSCC. MEDI0641 ablated CSC in the PDX-SCC-M0 model, reduced it by five-fold in the PDX-SCC-M1, and two-fold in the PDX-SCC-M11 model. Importantly, mice (n = 12) treated with neoadjuvant, single administration of MEDI0641 prior to surgical tumor removal showed no recurrence for more than 200 days, whereas the control group had 7 recurrences (in 12 mice; P = 0.0047).Conclusions: Collectively, these findings demonstrate that an anti-5T4 antibody-drug conjugate reduces the fraction of CSCs and prevents local recurrence and suggest a novel therapeutic approach for patients with HNSCC. Clin Cancer Res; 23(10); 2516-27. ©2016 AACR.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Immunoconjugates/administration & dosage , Membrane Glycoproteins/immunology , Animals , Benzodiazepines/administration & dosage , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Self Renewal/genetics , Cell Self Renewal/immunology , DNA Damage/drug effects , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/pathology , Humans , Immunoconjugates/immunology , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Pyrroles/administration & dosage , Squamous Cell Carcinoma of Head and Neck , Tissue Array Analysis , Treatment Outcome , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(34): 54120-54136, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27472462

ABSTRACT

Although endocrine therapy is successfully used to treat patients with estrogen receptor (ER) positive breast cancer, a substantial proportion of this population will relapse. Several mechanisms of acquired resistance have been described including activation of the mTOR pathway, increased activity of CDK4 and activating mutations in ER. Using a patient derived xenograft model harboring a common activating ER ligand binding domain mutation (D538G), we evaluated several combinatorial strategies using the selective estrogen receptor degrader (SERD) fulvestrant in combination with chromatin modifying agents, and CDK4/6 and mTOR inhibitors. In this model, fulvestrant binds WT and MT ER, reduces ER protein levels, and downregulated ER target gene expression. Addition of JQ1 or vorinostat to fulvestrant resulted in tumor regression (41% and 22% regression, respectively) though no efficacy was seen when either agent was given alone. Interestingly, although the CDK4/6 inhibitor palbociclib and mTOR inhibitor everolimus were efficacious as monotherapies, long-term delayed tumor growth was only observed when co-administered with fulvestrant. This observation was consistent with a greater inhibition of compensatory signaling when palbociclib and everolimus were co-dosed with fulvestrant. The addition of fulvestrant to JQ1, vorinostat, everolimus and palbociclib also significantly reduced lung metastatic burden as compared to monotherapy. The combination potential of fulvestrant with palbociclib or everolimus were confirmed in an MCF7 CRISPR model harboring the Y537S ER activating mutation. Taken together, these data suggest that fulvestrant may have an important role in the treatment of ER positive breast cancer with acquired ER mutations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Mutation , Receptors, Estrogen/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Everolimus/administration & dosage , Female , Fulvestrant , Humans , MCF-7 Cells , Mice , Piperazines/administration & dosage , Pyridines/administration & dosage , Receptors, Estrogen/analysis , Xenograft Model Antitumor Assays
12.
Cancer Res ; 76(16): 4661-72, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27325649

ABSTRACT

Dendritic cell (DC)-based vaccine strategies aimed at targeting cancer stem-like cells (CSC) may be most efficacious if deployed in the adjuvant setting. In this study, we offer preclinical evidence that this is the case for a CSC-DC vaccine as tested in murine models of SCC7 squamous cell cancer and D5 melanoma. Vaccination of mice with an ALDH(high) SCC7 CSC-DC vaccine after surgical excision of established SCC7 tumors reduced local tumor relapse and prolonged host survival. This effect was augmented significantly by simultaneous administration of anti-PD-L1, an immune checkpoint inhibitor. In the minimal disease setting of D5 melanoma, treatment of mice with ALDH(high) CSC-DC vaccination inhibited primary tumor growth, reduced spontaneous lung metastases, and increased host survival. In this setting, CCR10 and its ligands were downregulated on ALDH(high) D5 CSCs and in lung tissues, respectively, after vaccination with ALDH(high) D5 CSC-DC. RNAi-mediated attenuation of CCR10 blocked tumor cell migration in vitro and metastasis in vivo T cells harvested from mice vaccinated with ALDH(high) D5 CSC-DC selectively killed ALDH(high) D5 CSCs, with additional evidence of humoral immunologic engagement and a reduction in ALDH(high) cells in residual tumors. Overall, our results offered a preclinical proof of concept for the use of ALDH(high) CSC-DC vaccines in the adjuvant setting to more effectively limit local tumor recurrence and spontaneous pulmonary metastasis, as compared with traditional DC vaccines, with increased host survival further accentuated by simultaneous PD-L1 blockade. Cancer Res; 76(16); 4661-72. ©2016 AACR.


Subject(s)
Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Melanoma, Experimental , Neoplasms, Squamous Cell , Neoplastic Stem Cells/immunology , Adjuvants, Immunologic/pharmacology , Aldehyde Dehydrogenase/metabolism , Animals , Cancer Vaccines/immunology , Disease Models, Animal , Female , Flow Cytometry , Gene Knockdown Techniques , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
13.
Neoplasia ; 18(5): 273-281, 2016 05.
Article in English | MEDLINE | ID: mdl-27237319

ABSTRACT

Head and neck squamous cell carcinomas (HNSCC) exhibit a small population of uniquely tumorigenic cancer stem cells (CSC) endowed with self-renewal and multipotency. We have recently shown that IL-6 enhances the survival and tumorigenic potential of head and neck cancer stem cells (i.e. ALDH(high)CD44(high) cells). Here, we characterized the effect of therapeutic inhibition of IL-6 with a novel humanized anti-IL-6 antibody (MEDI5117) using three low-passage patient-derived xenograft (PDX) models of HNSCC. We observed that single agent MEDI5117 inhibited the growth of PDX-SCC-M1 tumors (P < .05). This PDX model was generated from a previously untreated HNSCC. In contrast, MEDI5117 was not effective at reducing overall tumor volume for PDX models representing resistant disease (PDX-SCC-M0, PDX-SCC-M11). Low dose MEDI5117 (3 mg/kg) consistently decreased the fraction of cancer stem cells in PDX models of HNSCC when compared to IgG-treated controls, as follows: PDX-SCC-M0 (P < .001), PDX-SCC-M1 (P < .001), PDX-SCC-M11 (P = .04). Interestingly, high dose MEDI5117 (30 mg/kg) decreased the CSC fraction in the PDX-SCC-M11 model (P = .002), but not in PDX-SCC-M0 and PDX-SCC-M1. MEDI5117 mediated a dose-dependent decrease in the number of orospheres generated by ALDH(high)CD44(high) cells cultured in ultra-low attachment plates (P < .05), supporting an inhibitory effect on head and neck cancer stem cells. Notably, single agent MEDI5117 reduced the overall recurrence rate of PDX-SCC-M0, a PDX generated from the local recurrence of human HNSCC. Collectively, these data demonstrate that therapeutic inhibition of IL-6 with low-dose MEDI5117 decreases the fraction of cancer stem cells, and that adjuvant MEDI5117 inhibits recurrence in preclinical models of HNSCC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Animals , Biomarkers , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Disease Models, Animal , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/mortality , Humans , Mice , Recurrence , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
14.
Cancer Res ; 76(2): 480-90, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26744529

ABSTRACT

Elevated levels of the proinflammatory cytokine IL6 are associated with poor survival outcomes in many cancers. Antibodies targeting IL6 and its receptor have been developed for chronic inflammatory disease, but they have not yet been shown to clearly benefit cancer patients, possibly due to antibody potency or the settings in which they have been tested. In this study, we describe the development of a novel high-affinity anti-IL6 antibody, MEDI5117, which features an extended half-life and potent inhibitory effects on IL6 biologic activity. MEDI5117 inhibited IL6-mediated activation of STAT3, suppressing the growth of several tumor types driven by IL6 autocrine signaling. In the same models, MEDI5117 displayed superior preclinical activity relative to a previously developed anti-IL6 antibody. Consistent with roles for IL6 in promoting tumor angiogenesis, we found that MEDI5117 inhibited the growth of endothelial cells, which can produce IL6 and support tumorigenesis. Notably, in tumor xenograft assays in mice, we documented the ability of MEDI5117 to enhance the antitumor activities of chemotherapy or gefitinib in combination treatment regimens. MEDI5117 also displayed robust activity on its own against trastuzumab-resistant HER2(+) tumor cells by targeting the CD44(+)CD24(-) cancer stem cell population. Collectively, our findings extend the evidence of important pleiotropic roles of IL6 in tumorigenesis and drug resistance, and offer a preclinical proof of concept for the use of IL6 antibodies in combination regimens to heighten therapeutic responses and overcome drug resistance.


Subject(s)
Interleukin-6/metabolism , Neoplasms/genetics , Trastuzumab/therapeutic use , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Neoplasms/drug therapy , Signal Transduction , Trastuzumab/administration & dosage
15.
Front Biosci (Elite Ed) ; 8(1): 181-8, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26709654

ABSTRACT

Cancer stem cells (CSCs) represent a rare subpopulation of tumor cells responsible for tumor initiation and eventual patient relapse. Strong preclinical evidence points to the importance of this population for therapeutic intervention and currently there are several anti-CSC therapies under development. While we begin to advance these therapies into the clinic, there is a need to develop biomarkers to measure both patient response and prognosis. This review will discuss the experimental evidence for the role of CSCs in patient outcomes as well as the need, challenges and the hope that CSC biomarkers may provide.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplastic Stem Cells/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy
16.
World J Stem Cells ; 7(1): 27-36, 2015 Jan 26.
Article in English | MEDLINE | ID: mdl-25621103

ABSTRACT

The origins of the complex process of intratumoral heterogeneity have been highly debated and different cellular mechanisms have been hypothesized to account for the diversity within a tumor. The clonal evolution and cancer stem cell (CSC) models have been proposed as drivers of this heterogeneity. However, the concept of cancer stem cell plasticity and bidirectional conversion between stem and non-stem cells has added additional complexity to these highly studied paradigms and may help explain the tumor heterogeneity observed in solid tumors. The process of cancer stem cell plasticity in which cancer cells harbor the dynamic ability of shifting from a non-CSC state to a CSC state and vice versa may be modulated by specific microenvironmental signals and cellular interactions arising in the tumor niche. In addition to promoting CSC plasticity, these interactions may contribute to the cellular transformation of tumor cells and affect response to chemotherapeutic and radiation treatments by providing CSCs protection from these agents. Herein, we review the literature in support of this dynamic CSC state, discuss the effectors of plasticity, and examine their role in the development and treatment of cancer.

17.
Mol Cancer Ther ; 13(2): 386-98, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344235

ABSTRACT

The hedgehog pathway has been implicated in the tumorigenesis, tumor progression, and metastasis of numerous human cancers. We generated the first fully human hedgehog antibody MEDI-5304 and characterized its antitumor activity and preclinical toxicology. MEDI-5304 bound sonic hedgehog (SHH) and Indian hedgehog (IHH) with low picomolar affinity and neutralized SHH and IHH activity in cellular mGLI1 reporter assays. The antibody inhibited transcription of hedgehog target genes and osteoblast differentiation of C3H10T1/2 cells. We evaluated the activity of MEDI-5304 in vivo in model systems that allowed us to evaluate two primary hypotheses of hedgehog function in human cancer, paracrine signaling between tumor and stromal cells and cancer stem cell (CSC) self-renewal. MEDI-5304 displayed robust pharmacodynamic effects in stromal cells that translated to antitumor efficacy as a single agent in an HT-29/MEF coimplantation model of paracrine hedgehog signaling. MEDI-5304 also improved responses to carboplatin in the HT-29/MEF model. The antibody, however, had no effect as a single agent or in combination with gemcitabine on the CSC frequency or growth of several primary pancreatic cancer explant models. These findings support the conclusion that hedgehog contributes to tumor biology via paracrine tumor-stromal signaling but not via CSC maintenance or propagation. Finally, the only safety study finding associated with MEDI-5304 was ondontodysplasia in rats. Thus, MEDI-5304 represents a potent dual hedgehog inhibitor suitable for continued development to evaluate efficacy and safety in human patients with tumors harboring elevated levels of SHH or IHH.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Paracrine Communication/drug effects , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , HT29 Cells , Hedgehog Proteins/immunology , Humans , Kinetics , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , NIH 3T3 Cells , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Paracrine Communication/immunology , Protein Binding/immunology , Rats, Wistar , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
18.
Stem Cells Transl Med ; 2(1): 43-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23283488

ABSTRACT

Although cancer is largely seen as a disease stemming from genetic mutations, evidence has implicated epigenetic regulation of gene expression as a driving force for tumorigenesis. Epigenetic regulation by histone modification, specifically through polycomb group (PcG) proteins such as EZH2 and BMI-1, is a major driver in stem cell biology and is found to be correlated with poor prognosis in many tumor types. This suggests a role for PcG proteins in cancer stem cells (CSCs). We hypothesized that epigenetic modification by EZH2, specifically, helps maintain the CSC phenotype and that in turn this epigenetic modifier can be used as a reporter for CSC activity in an in vitro high-throughput screening assay. CSCs isolated from pancreatic and breast cancer lines had elevated EZH2 levels over non-CSCs. Moreover, EZH2 knockdown by RNA interference significantly reduced the frequency of CSCs in all models tested, confirming the role of EZH2 in maintenance of the CSC population. Interestingly, genes affected by EZH2 loss, and therefore CSC loss, were inversely correlated with genes identified by CSC enrichment, further supporting the function of EZH2 CSC regulation. We translated these results into a novel assay whereby elevated EZH2 staining was used as a reporter for CSCs. Data confirmed that this assay could effectively measure changes, both inhibition and enrichment, in the CSC population, providing a novel approach to look at CSC activity. This assay provides a unique, rapid way to facilitate CSC screening across several tumor types to aid in further CSC-related research.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , Biomarkers, Tumor/genetics , Breast Neoplasms , Cell Line, Tumor , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histones/metabolism , Humans , Neoplastic Stem Cells/physiology , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms , Polycomb Repressive Complex 2/genetics , RNA Interference , Transcriptome
19.
J Bone Marrow Res ; 1: 122, 2013 May 23.
Article in English | MEDLINE | ID: mdl-24772452

ABSTRACT

BACKGROUND: Human bone marrow derived mesenchymal stem cells (hMSCs) are capable of differentiation into multiple cell lineages and demonstrate a wide variety of use in various therapeutic applications. Only recently has research begun to understand the gene expression profiles of hMSCs and their differentiated counterparts in vivo and ex vivo. PURPOSE: The research presented here aimed at gaining a better understanding of gene expression patterns present during hMSC invasion through a basement membrane. METHODS: Changes in gene expression were evaluated between invasive and non-invasive cells using Agilent's gene expression arrays and Matrigel invasion chambers. The cells were specifically attracted to a defined stem cell media called SCM. RESULTS: A total 435 genes were up-regulated by 2- fold or more in the invasive population of cells and classified into developmental programs and immunological/inflammatory signaling pathways determined by Ingenuity Pathway Analysis (IPA). This list included a variety of regulators of growth and differentiation including NANOG, STAT3 and STAT5A and members of the polycomb repressive complex-2 (PCRC2) EZH2 and SUZ12. The known regulator of inflammation and hypoxia HIF-1α was also increased suggesting that regulation of the microenvironment is important during this process. Finally, the invasion process could be reversed using the STAT3 inhibitor Static. CONCLUSIONS: Overall these data will increase the understanding of the genetic pathways functioning during hMSC invasion and aid in the development of their therapeutic applications.

20.
J Mol Med (Berl) ; 90(7): 791-801, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22660276

ABSTRACT

Normal development and homeostasis requires a carefully coordinated gene expression program. Appropriate transcriptional regulation is maintained, in part, through epigenetic modifications of both DNA and histones. It is now apparent that the epigenetic landscape is complex and carefully controlled to both silence and activate gene transcription and that these states remain exquisitely poised for reversal. The deregulation of epigenetics in cancer is common and results in both the activation of oncogenes and the silencing of tumor suppressors. A tremendous amount of research corroborates the existence in many tumor types of a cancer stem cell that is both the origin and cell type responsible for resistance of tumors to current therapies. As our understanding of cancer stem cell biology continues, it is apparent that these cells are also under the influence of epigenetic regulation. We will discuss the cancer stem cell hypothesis and the role of epigenetics in both normal and cancer stem cell biology.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Stem Cells/metabolism , Animals , DNA Methylation , Humans , Neoplasms/metabolism , Neoplasms/therapy , Neoplastic Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL