Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Front Cell Dev Biol ; 11: 1270398, 2023.
Article in English | MEDLINE | ID: mdl-38020903

ABSTRACT

Introduction: The glycoengineered type II anti-CD20 monoclonal antibody obinutuzumab has been licensed for treatment in follicular non-Hodgkin lymphoma and B-CLL following clinical trials demonstrating superior outcomes to standard of care treatment. However, ultimately many patients still relapse, highlighting the need to understand the mechanisms behind treatment failure to improve patient care. Resistance to chemotherapy is often caused by the ability of malignant B-cells to migrate to the bone marrow and home into the stromal layer. Therefore, this study aimed to investigate whether stromal cells were also able to inhibit type II anti-CD20 antibody mechanisms of action, contributing to resistance to therapy. Methods: A stromal-tumor co-culture was established in vitro between Raji or Daudi B-cell tumor cells and M210B4 stromal cells in 24 well plates. Results: Contact with stromal cells was able to protect tumor cells from obinutuzumab mediated programmed cell death (PCD), antibody dependent cellular phagocytosis and antibody dependent cellular cytotoxicity. Furthermore, such protection required direct contact between stroma and tumor cells. Stromal cells appeared to interfere with obinutuzumab mediated B-cell homotypic adhesion through inhibiting and reversing actin remodelling, potentially as a result of stromal-tumor cell contact leading to downregulation of CD20 on the surface of tumor cells. Further evidence for the potential role of CD20 downregulation comes through the reduction in surface CD20 expression and inhibition of obinutuzumab mediated PCD when tumor cells are treated with Ibrutinib in the presence of stromal cells. The proteomic analysis of tumor cells after contact with stromal cells led to the identification of a number of altered pathways including those involved in cell adhesion and the actin cytoskeleton and remodeling. Discussion: This work demonstrates that contact between tumor cells and stromal cells leads to inhibition of Obinutuzumab effector functions and has important implications for future therapies to improve outcomes to anti-CD20 antibodies. A deeper understanding of how anti-CD20 antibodies interact with stromal cells could prove a useful tool to define better strategies to target the micro-environment and ultimately improve patient outcomes in B-cell malignancies.

2.
Oncoimmunology ; 12(1): 2223094, 2023.
Article in English | MEDLINE | ID: mdl-37332616

ABSTRACT

Despite breakthroughs in immune checkpoint inhibitors (ICI), the majority of tumors, including those poorly infiltrated by CD8+ T cells or heavily infiltrated by immunosuppressive immune effector cells, are unlikely to result in clinically meaningful tumor responses. Radiation therapy (RT) has been combined with ICI to potentially overcome this resistance and improve response rates but reported clinical trial results have thus far been disappointing. Novel approaches are required to overcome this resistance and reprogram the immunosuppressive tumor microenvironment (TME) and address this major unmet clinical need. Using diverse preclinical tumor models of prostate and bladder cancer, including an autochthonous prostate tumor (Pten-/-/trp53-/-) that respond poorly to radiation therapy (RT) and anti-PD-L1 combinations, the key drivers of this resistance within the TME were profiled and used to develop rationalized combination therapies that simultaneously enhance activation of anti-cancer T cell responses and reprogram the immunosuppressive TME. The addition of anti-CD40mAb to RT resulted in an increase in IFN-y signaling, activation of Th-1 pathways with an increased infiltration of CD8+ T-cells and regulatory T-cells with associated activation of the CTLA-4 signaling pathway in the TME. Anti-CTLA-4mAb in combination with RT further reprogrammed the immunosuppressive TME, resulting in durable, long-term tumor control. Our data provide novel insights into the underlying mechanisms of the immunosuppressive TME that result in resistance to RT and anti-PD-1 inhibitors and inform therapeutic approaches to reprogramming the immune contexture in the TME to potentially improve tumor responses and clinical outcomes.


Subject(s)
Tumor Microenvironment , Urinary Bladder Neoplasms , Male , Humans , T-Lymphocytes, Regulatory/metabolism , Signal Transduction , Combined Modality Therapy , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/radiotherapy
3.
J Clin Oncol ; 40(20): 2203-2212, 2022 07 10.
Article in English | MEDLINE | ID: mdl-35385334

ABSTRACT

PURPOSE: There is a need to refine the selection of patients with oropharyngeal squamous cell carcinoma (OPSCC) for treatment de-escalation. We investigated whether pretreatment absolute lymphocyte count (ALC) predicted overall survival (OS) benefit from the addition of concurrent chemotherapy to radical radiotherapy. PATIENTS AND METHODS: This was an observational study of consecutive OPSCCs treated by curative-intent radiotherapy, with or without concurrent chemotherapy (n = 791) with external, independent validation from a separate institution (n = 609). The primary end point was OS at 5 years. Locoregional control (LRC) was assessed using competing risk regression as a secondary end point. Previously determined prognostic factors were used in a multivariable Cox proportional hazards model to assess the prognostic importance of ALC and the interaction between ALC and cisplatin chemotherapy use. RESULTS: Pretreatment ALC was prognostic for 5-year OS on multivariable analysis (hazard ratio [HR] 0.64; 95% CI, 0.42 to 0.98; P = .04). It also predicted benefit from the use of concurrent cisplatin chemotherapy, with a significant interaction between cisplatin chemotherapy and pretreatment ALC (likelihood ratio test, P = .04): higher ALC count reduced the 5-year OS benefit compared with radiotherapy alone (HR 2.53; 95% CI, 1.03 to 6.19; P = .043). This was likely driven by an effect on LRC up to 5 years (interaction subdistribution HR 2.29; 95% CI, 0.68 to 7.71; P = .094). An independent validation cohort replicated the OS (HR 2.53; 95% CI, 0.98 to 6.52; P = .055) and LRC findings (interaction subdistribution HR 3.43; 95% CI, 1.23 to 9.52; P = .018). CONCLUSION: For OPSCC, the pretreatment ALC is prognostic for OS and also predicts benefit from the addition of cisplatin chemotherapy to radiotherapy. These findings require prospective evaluation, and could inform the selection of good prognosis patients for a de-escalation trial.


Subject(s)
Cisplatin , Oropharyngeal Neoplasms , Disease-Free Survival , Humans , Lymphocyte Count , Oropharyngeal Neoplasms/drug therapy , Oropharyngeal Neoplasms/radiotherapy , Prognosis , Proportional Hazards Models
4.
Radiat Oncol ; 15(1): 254, 2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33148287

ABSTRACT

Radiotherapy (RT) is a highly effective anti-cancer therapy delivered to around 50-60% of patients. It is part of therapy for around 40% of cancer patients who are cured of their disease. Until recently, the focus of this anti-tumour efficacy has been on the direct tumour cytotoxicity and RT-induced DNA damage. Recently, the immunomodulatory effects of RT on the tumour microenvironment have increasingly been recognized. There is now intense interest in potentially using RT to induce an anti-tumour immune response, which has led to rethinking into how the efficacy of RT could be further enhanced. Following the breakthrough of immune check point inhibitors (ICIs), a new era of immuno-oncology (IO) agents has emerged and established immunotherapy as a routine part of cancer treatment. Despite ICI improving outcomes in many cancer types, overall durable responses occur in only a minority of patients. The immunostimulatory effects of RT make combinations with ICI attractive to potentially amplify anti-tumour immunity resulting in increased tumour responses and improved outcomes. In contrast, tumours with profoundly immunosuppressive tumour microenvironments, dominated by myeloid-derived cell populations, remain a greater clinical challenge and RT may potentially further enhance the immunosuppression. To harness the full potential of RT and IO agent combinations, further insights are required to enhance our understanding of the role these immunosuppressive myeloid populations play, how RT influences these populations and how they may be therapeutically manipulated in combination with RT to improve outcomes further. These are exciting times with increasing numbers of IO targets being discovered and IO agents undergoing clinical evaluation. Multidisciplinary research collaborations will be required to establish the optimal parameters for delivering RT (target volume, dose and fractionation) in combination with IO agents, including scheduling to achieve maximal therapeutic efficacy.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/radiotherapy , Tumor Microenvironment , Combined Modality Therapy , Humans , Immunotherapy , Neoplasms/immunology , T-Lymphocytes/physiology
5.
Cancers (Basel) ; 12(10)2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33008040

ABSTRACT

Radiotherapy (RT) is a highly effective anti-cancer treatment. Immunotherapy using immune checkpoint blockade (ICI) has emerged as a new and robust pillar in cancer therapy; however, the response rate to single agent ICI is low whilst toxicity remains. Radiotherapy has been shown to have local and systemic immunomodulatory effects. Therefore, combining RT and immunotherapy is a rational approach to enhance anti-tumour immune responses. However, the immunomodulatory effects of RT can be both immunostimulatory or immunosuppressive and may be different across different tumour types and patients. Therefore, there is an urgent medical need to establish biomarkers to guide clinical decision making in predicting responses or in patient selection for RT-based combination treatments. In this review, we summarize the immunological effects of RT on the tumour microenvironment and emerging biomarkers to help better understand the implications of these immunological changes, and we provide new insights into the potential for combination therapies with RT and immunotherapy.

6.
J Exp Med ; 216(5): 1038-1049, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30936262

ABSTRACT

CAR T cells represent a potentially curative strategy for B cell malignancies. However, the outcome and dynamics of CAR T cell interactions in distinct anatomical sites are poorly understood. Using intravital imaging, we tracked interactions established by anti-CD19 CAR T cells in B cell lymphoma-bearing mice. Circulating targets trapped CAR T cells in the lungs, reducing their access to lymphoid organs. In the bone marrow, tumor apoptosis was largely due to CAR T cells that engaged, killed, and detached from their targets within 25 min. Notably, not all CAR T cell contacts elicited calcium signaling or killing while interacting with tumors, uncovering extensive functional heterogeneity. Mathematical modeling revealed that direct killing was sufficient for tumor regression. Finally, antigen-loss variants emerged in the bone marrow, but not in lymph nodes, where CAR T cell cytotoxic activity was reduced. Our results identify a previously unappreciated level of diversity in the outcomes of CAR T cell interactions in vivo, with important clinical implications.


Subject(s)
Immunotherapy, Adoptive/methods , Intravital Microscopy/methods , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/metabolism , Single-Cell Analysis/methods , T-Lymphocytes/metabolism , Animals , Antigens, CD19/metabolism , Apoptosis , Cell Line, Tumor , Lung/metabolism , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Models, Theoretical , Recurrence
7.
Cancer Immunol Immunother ; 67(10): 1519-1531, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30039427

ABSTRACT

Epithelial ovarian cancer (EOC) is the leading cause of gynaecological cancer-related death in Europe. Although most patients achieve an initial complete response with first-line treatment, recurrence occurs in more than 80% of cases. Thus, there is a clear unmet need for novel second-line treatments. EOC is frequently infiltrated with T lymphocytes, the presence of which has been shown to be associated with improved clinical outcomes. Adoptive T-cell therapy (ACT) using ex vivo-expanded tumour-infiltrating lymphocytes (TILs) has shown remarkable efficacy in other immunogenic tumours, and may represent a promising therapeutic strategy for EOC. In this preclinical study, we investigated the efficacy of using anti-CD3/anti-CD28 magnetic beads and IL-2 to expand TILs from freshly resected ovarian tumours. TILs were expanded for up to 3 weeks, and then subjected to a rapid-expansion protocol (REP) using irradiated feeder cells. Tumours were collected from 45 patients with EOC and TILs were successfully expanded from 89.7% of biopsies. Expanded CD4+ and CD8+ subsets demonstrated features associated with memory phenotypes, and had significantly higher expression of key activation and functional markers than unexpanded TILs. Expanded TILs produced anti-tumour cytokines when co-cultured with autologous tumour cells, inferring tumour cytotoxicity. Our findings demonstrate that it is possible to re-activate and expand tumour-reactive T cells from ovarian tumours. This presents a promising immunotherapy that could be used sequentially or in combination with current therapeutic strategies.


Subject(s)
Adenocarcinoma, Clear Cell/therapy , Carcinosarcoma/therapy , Cystadenocarcinoma, Serous/therapy , Cytokines/metabolism , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/therapy , Adenocarcinoma, Clear Cell/immunology , Adenocarcinoma, Clear Cell/metabolism , Aged , Carcinosarcoma/immunology , Carcinosarcoma/metabolism , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/metabolism , Cytotoxicity, Immunologic , Female , Humans , Middle Aged , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured
8.
J Immunother ; 41(3): 130-140, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29239915

ABSTRACT

Chimeric antigen receptor (CAR) T cells represent a novel targeted approach to overcome both quantitative and qualitative shortfalls of the host immune system relating to the detection and subsequent destruction of tumors. The identification of antigens expressed specifically on the surface of tumor cells is a critical first step in the ability to utilize CAR T cells for the treatment of cancer. The 5T4 is a tumor-associated antigen which is expressed on the cell surface of most solid tumors including ovarian cancer. Matched blood and tumor samples were collected from 12 patients with ovarian cancer; all tumors were positive for 5T4 expression by immunohistochemistry. Patient T cells were effectively transduced with 2 different anti-5T4 CAR constructs which differed in their affinity for the target antigen. Co-culture of CAR T cells with matched autologous tumor disaggregates resulted in antigen-specific secretion of IFN-gamma. Furthermore, assessment of the efficacy of anti-5T4 CAR T cells in a mouse model resulted in therapeutic benefit against established ovarian tumors. These results demonstrate proof of principle that 5T4 is an attractive target for immune intervention in ovarian cancer and that patient T cells engineered to express a 5T4-specific CAR can recognize and respond physiologically to autologous tumor cells.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy, Adoptive , Membrane Glycoproteins/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression , Humans , Immunotherapy, Adoptive/methods , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, Chimeric Antigen/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 23(18): 5514-5526, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28533222

ABSTRACT

Purpose: Radiotherapy is a highly effective anticancer treatment forming part of the standard of care for the majority of patients, but local and distal disease recurrence remains a major cause of mortality. Radiotherapy is known to enhance tumor immunogenicity; however, the contribution and mechanisms of radiotherapy-induced immune responses are unknown.Experimental Design: The impact of low-dose fractionated radiotherapy (5 × 2 Gy) alone and in combination with αPD-1 mAb on the tumor microenvironment was evaluated by flow cytometry and next-generation sequencing of the T-cell receptor (TCR) repertoire. A dual-tumor model was used, with fractionated radiotherapy delivered to a single tumor site to enable evaluation of the local and systemic response to treatment and ability to induce abscopal responses outside the radiation field.Results: We show that fractionated radiotherapy leads to T-cell infiltration at the irradiated site; however, the TCR landscape remains dominated by polyclonal expansion of preexisting T-cell clones. Adaptive resistance via the PD-1/PD-L1 pathway restricts the generation of systemic anticancer immunity following radiotherapy, which can be overcome through combination with αPD-1 mAb leading to improved local and distal tumor control. Moreover, we show that effective clearance of tumor following combination therapy is dependent on both T cells resident in the tumor at the time of radiotherapy and infiltrating T cells.Conclusions: These data provide evidence that radiotherapy can enhance T-cell trafficking to locally treated tumor sites and augment preexisting anticancer T-cell responses with the capacity to mediate regression of out-of-field tumor lesions when delivered in combination with αPD-1 mAb therapy. Clin Cancer Res; 23(18); 5514-26. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/radiation effects , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/radiation effects , Animals , Cell Line, Tumor , Combined Modality Therapy , Cytokines/metabolism , Disease Models, Animal , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/metabolism , Neoplasms/therapy , Programmed Cell Death 1 Receptor/metabolism , Radiotherapy , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Survival Rate , T-Lymphocyte Subsets/metabolism , Xenograft Model Antitumor Assays
10.
Cancer Immunol Res ; 4(7): 621-630, 2016 07.
Article in English | MEDLINE | ID: mdl-27241845

ABSTRACT

Tumor cells dying after cytotoxic therapy are a potential source of antigen for T-cell priming. Antigen-presenting cells (APC) can cross-present MHC I-restricted peptides after the uptake of dying cells. Depending on the nature of the surrounding environmental signals, APCs then orchestrate a spectrum of responses ranging from immune activation to inhibition. Previously, we had demonstrated that combining radiation with either agonistic monoclonal antibody (mAb) to CD40 or a systemically administered TLR7 agonist could enhance CD8 T-cell-dependent protection against syngeneic murine lymphoma models. However, it remains unknown how individual APC populations affect this antitumor immune response. Using APC depletion models, we now show that dendritic cells (DC), but not macrophages or B cells, were responsible for the generation of long-term immunologic protection following combination therapy with radiotherapy and either agonistic CD40 mAb or systemic TLR7 agonist therapy. Novel immunotherapeutic approaches that augment antigen uptake and presentation by DCs may further enhance the generation of therapeutic antitumor immune responses, leading to improved outcomes after radiotherapy. Cancer Immunol Res; 4(7); 621-30. ©2016 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunotherapy , Lymphocyte Activation/immunology , Neoplasms/immunology , Radiotherapy , Animals , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/radiation effects , Combined Modality Therapy , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/radiation effects , Disease Models, Animal , Lymphocyte Depletion , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/radiation effects , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Phagocytosis
11.
Leuk Lymphoma ; 57(6): 1269-80, 2016.
Article in English | MEDLINE | ID: mdl-27050042

ABSTRACT

Nearly two decades ago rituximab heralded a new era in management of B cell malignancies significantly increasing response rates and survival. However, despite clear therapeutic advantage, significant numbers of patients become refractory to anti-CD20 mAb therapy, suggesting urgent improvements are required. It is now well recognized that the suppressive tumor microenvironment plays an important role in the outcome of anti-CD20 mAb therapy and that manipulation of this environment may improve the efficacy and produce long-term tumor control. The past few years have seen a surge of interest in immunomodulatory agents capable of overwriting immune suppressive networks into favorable clinical outcome. Currently, a number of such combinations with anti-CD20 mAb is under evaluation and some have produced encouraging outcomes in rituximab refractory disease. In this review, we give an outline of anti-CD20 mAbs and explore the combinations with immunomodulatory agents that enhance antitumor immunity through targeting stimulatory or inhibitory pathways and have proven potential to synergize with anti-CD20 mAb therapy. These agents, primarily mAbs, target CTLA-4, PD-1/PD-L1, and CD40.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunologic Factors/therapeutic use , Immunomodulation/drug effects , Leukemia, B-Cell/drug therapy , Lymphoma, B-Cell/drug therapy , Rituximab/therapeutic use , Animals , Antigens, CD20 , Antineoplastic Agents/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD40 Antigens/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Cytotoxicity, Immunologic/drug effects , Humans , Immunologic Factors/pharmacology , Leukemia, B-Cell/immunology , Leukemia, B-Cell/metabolism , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Rituximab/pharmacology , Signal Transduction/drug effects
12.
Oncotarget ; 7(13): 17035-46, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26959743

ABSTRACT

Strategies to augment anti-cancer immune responses have recently demonstrated therapeutic utility. To date clinical success has been achieved through targeting co-inhibitory checkpoints such as CTLA-4, PD-1, and PD-L1. However, approaches that target co-activatory pathways are also being actively being developed. Here we report that the novel TLR7-selective agonist DSR-29133 is well tolerated in mice and leads to acute immune activation. Administration of DSR-29133 leads to the induction of IFNα/γ, IP-10, TNFα, IL-1Ra and IL-12p70, and to a reduction in tumor burden in syngeneic models of renal cancer (Renca), metastatic osteosarcoma (LM8) and colorectal cancer (CT26). Moreover, we show that the efficacy of DSR-29133 was significantly improved when administered in combination with low-dose fractionated radiotherapy (RT). Effective combination therapy required weekly administration of DSR-29133 commencing on day 1 of a fractionated RT treatment cycle, whereas no enhancement of radiation response was observed when DSR-29133 was administered at the end of the fractionated RT cycle. Combined therapy resulted in curative responses in a high proportion of mice bearing established CT26 tumors which was dependent on the activity of CD8+ T-cells but independent of CD4+ T-cells and NK/NKT cells. Moreover, long-term surviving mice originally treated with DSR-29133 and RT were protected by a tumor-specific memory immune response which could prevent tumor growth upon rechallenge. These results demonstrate that DSR-29133 is a potent selective TLR7 agonist that when administered intravenously can induce anti-tumor immune responses that can be further enhanced through combination with low-dose fractionated RT.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Chemoradiotherapy/methods , Neoplasms, Experimental/drug therapy , Toll-Like Receptor 7/agonists , Adenine/pharmacology , Administration, Intravenous , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Dose Fractionation, Radiation , Humans , Lymphocyte Activation/drug effects , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/radiotherapy
13.
Expert Opin Biol Ther ; 15(6): 787-801, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25882106

ABSTRACT

INTRODUCTION: After years of limited success, progress of anti-cancer immuno-therapeutics has been considerable over the past decade. Key to this progress has been the application of new biological insights around the importance and nature of immune checkpoints that are able to reverse down-regulation of anti-tumor immunity. AREAS COVERED: An overview of the preclinical and recent clinical trial data on key immuno-regulatory agents currently in development, including antibody targeting of cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed death receptor 1 (PD-1) on T-lymphocytes and its principal ligand (PD-L1) on tumor cells as well as immune agonists (e.g., anti-CD40). EXPERT OPINION: Durable long-term responses in some patients with advanced melanoma, initially with ipilimumab (anti-CTLA-4) and more recently antibodies targeting either PD-1 or PD-L1 in patients with melanoma and renal cancer, non-small-cell lung, bladder and head and neck cancers with less toxicity, have provided real optimism that immunotherapeutic approaches can improve outcomes in a wide range of cancer. The manageable tolerability of PD-1-pathway blockers and their unique mechanism of action are encouraging combination approaches. Current efforts focus on registration trials of single agents plus combinations in many different tumor types and treatment settings and identifying and developing predictive biomarkers of immunological response.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Immunotherapy/trends , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neoplasm/administration & dosage , CTLA-4 Antigen/immunology , Humans , Ipilimumab , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Treatment Outcome
14.
Cancer Res ; 74(19): 5458-68, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25274032

ABSTRACT

Radiotherapy is a major part in the treatment of most common cancers, but many patients experience local recurrence with metastatic disease. In evaluating response biomarkers, we found that low doses of fractionated radiotherapy led to PD-L1 upregulation on tumor cells in a variety of syngeneic mouse models of cancer. Notably, fractionated radiotherapy delivered in combination with αPD-1 or αPD-L1 mAbs generated efficacious CD8(+) T-cell responses that improved local tumor control, long-term survival, and protection against tumor rechallenge. These favorable outcomes were associated with induction of a tumor antigen-specific memory immune response. Mechanistic investigations showed that IFNγ produced by CD8(+) T cells was responsible for mediating PD-L1 upregulation on tumor cells after delivery of fractionated radiotherapy. Scheduling of anti-PD-L1 mAb was important for therapeutic outcome, with concomitant but not sequential administration with fractionated radiotherapy required to improve survival. Taken together, our results reveal the mechanistic basis for an adaptive response by tumor cells that mediates resistance to fractionated radiotherapy and its treatment failure. With attention to scheduling, combination immunoradiotherapy with radiotherapy and PD-1/PD-L1 signaling blockade may offer an immediate strategy for clinical evaluation to improve treatment outcomes.


Subject(s)
B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Dose Fractionation, Radiation , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Disease Models, Animal , Immunologic Memory , Mice
15.
Expert Opin Biol Ther ; 14(10): 1507-17, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25190612

ABSTRACT

INTRODUCTION: The anti-CD20 mAb rituximab has revolutionized the treatment of B-cell malignancies, improving outcome for patients. Despite these improvements, the majority of patients still relapse and become refractory to rituximab. Further efforts to improve anti-CD20 mAb efficacy have recently focused on obinutuzumab /GA101, a novel anti-CD20 mAb glycoengineered to display enhanced Fc-mediated effector mechanisms and induce direct cell death. AREAS COVERED: We provide an overview of the current insights into the mechanisms of action of obinutuzumab focusing on how structural modifications and differences to rituximab led to designation of obinutuzumab as a type II antibody. We summarize data from preclinical studies and recent clinical trials including the Phase III trial in chronic lymphocytic leukemia (CLL), which led to FDA approval in November 2013. EXPERT OPINION: Clinical data are now emerging confirming the promise of the initial preclinical data that demonstrated superior efficacy of obinutuzumab over rituximab at similar dosing. The emerging randomized Phase III data from older comorbid patients with previously untreated CLL demonstrated significant improvements in molecular remission rates and median progression-free survival of obinutuzumab plus chlorambucil versus rituximab plus chlorambucil. This emerging data provide reasons to be optimistic that outcomes for patients with B-cell malignancies can be further improved with obinutuzumab.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Humans , Treatment Outcome
16.
J Immunol ; 192(8): 3654-65, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24623129

ABSTRACT

T cells engrafted with chimeric AgRs (CAR) are showing exciting potential for targeting B cell malignancies in early-phase clinical trials. To determine whether the second-generation CAR was essential for optimal antitumor activity, two CD28-based CAR constructs targeting CD19 were tested for their ability to redirect mouse T cell function against established B cell lymphoma in a BALB/c syngeneic model system. T cells armed with either CAR eliminated A20 B cell lymphoma in vivo; however, one construct induced a T cell dose-dependent acute toxicity associated with a raised serum Th1 type cytokine profile on transfer into preconditioned mice. Moreover, a chronic toxicity manifested as granuloma-like formation in spleen, liver, and lymph nodes was observed in animals receiving T cells bearing either CD28 CAR, albeit with different kinetics dependent upon the specific receptor used. This phenotype was associated with an expansion of CD4+ CAR+ T cells and CD11b+ Gr-1(+) myeloid cells and increased serum Th2-type cytokines, including IL-10 and IL-13. Mouse T cells engrafted with a first-generation CAR failed to develop such autotoxicity, whereas toxicity was not apparent when T cells bearing the same receptors were transferred into C57BL/6 or C3H animals. In summary, the adoptive transfer of second-generation CD19-specific CAR T cells can result in a cell dose-dependent acute toxicity, whereas the prolonged secretion of high levels of Th2 cytokines from these CAR T cells in vivo drives a granulomatous reaction resulting in chronic toxicity. Strategies that prevent a prolonged Th2-cytokine biased CAR T cell response are clearly warranted.


Subject(s)
Antigens, CD19/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adoptive Transfer/adverse effects , Animals , Antigens, CD19/genetics , Cluster Analysis , Gene Expression Profiling , Humans , Immunophenotyping , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , Phenotype , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/transplantation , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
17.
Cancer Immunol Immunother ; 63(2): 133-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24190544

ABSTRACT

Adoptive cell therapy employing gene-modified T-cells expressing chimeric antigen receptors (CARs) has shown promising preclinical activity in a range of model systems and is now being tested in the clinical setting. The manufacture of CAR T-cells requires compliance with national and European regulations for the production of medicinal products. We established such a compliant process to produce T-cells armed with a first-generation CAR specific for carcinoembryonic antigen (CEA). CAR T-cells were successfully generated for 14 patients with advanced CEA(+) malignancy. Of note, in the majority of patients, the defined procedure generated predominantly CD4(+) CAR T-cells with the general T-cell population bearing an effector-memory phenotype and high in vitro effector function. Thus, improving the process to generate less-differentiated T-cells would be more desirable in the future for effective adoptive gene-modified T-cell therapy. However, these results confirm that CAR T-cells can be generated in a manner compliant with regulations governing medicinal products in the European Union.


Subject(s)
Adoptive Transfer , Carcinoembryonic Antigen/immunology , Chimerin Proteins/biosynthesis , Receptors, Antigen, T-Cell/biosynthesis , T-Lymphocytes/immunology , Humans , Immunophenotyping , Interferon-gamma/biosynthesis
18.
Immunol Rev ; 257(1): 91-106, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24329792

ABSTRACT

Blockbuster antibody therapies have catapulted immune-based approaches to treat cancer into the consciousness of mainstay clinical research. On the back of this, other emerging immune-based therapies are providing great promise. T-cell therapy is one such area where recent trials using T cells genetically modified to express an antibody-based chimeric antigen receptor (CAR) targeted against the CD19 antigen have demonstrated impressive responses when adoptively transferred to patients with advanced chronic lymphocytic leukemia. The general concept of the CAR T cell was devised some 20 years ago. In this relatively short period of time, the technology to redirect T-cell function has moved at pace facilitating clinical translation; however, many questions remain with respect to developing the approach to improve CAR T-cell therapeutic activity and also to broaden the range of tumors that can be effectively targeted by this approach. This review highlights some of the underlying principles and compromises of CAR T-cell technology using the CD19-targeted CAR as a paradigm and discusses some of the issues that relate to targeting solid tumors with CAR T cells.


Subject(s)
Antigens, CD19/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Culture Techniques , Gene Transfer Techniques , Genetic Engineering , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/immunology , Leukemia, B-Cell/therapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy
20.
Immunotherapy ; 5(9): 927-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23998728

ABSTRACT

Adoptive therapy using gene-modified T cells to express chimeric antigen receptors (CARs) is gaining pace in the clinic, particularly in hematological malignancies. Translation into treatment of solid tumors has been slower, not least because of the lack of truly tumor-specific target antigens. Alonso-Camino et al. describe experiments that further develop the concept of using the therapeutic entity (in this case, the CAR T cell) to screen for functional binding of tumor target cells. This article highlights the potential for the approach, but also underlies some of the key hurdles that remain to be overcome in order to produce a functional antibody-based screening approach that is able to identify novel tumor antigens that can be recognized by CAR T cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...