151
|
Combination immunotherapies implementing adoptive T-cell transfer for advanced-stage melanoma. Melanoma Res 2019. [PMID: 29521881 DOI: 10.1097/cmr.0000000000000436] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunotherapy is a promising method of treatment for a number of cancers. Many of the curative results have been seen specifically in advanced-stage melanoma. Despite this, single-agent therapies are only successful in a small percentage of patients, and relapse is very common. As chemotherapy is becoming a thing of the past for treatment of melanoma, the combination of cellular therapies with immunotherapies appears to be on the rise in in-vivo models and in clinical trials. These forms of therapies include tumor-infiltrating lymphocytes, T-cell receptor, or chimeric antigen receptor-modified T cells, cytokines [interleukin (IL-2), IL-15, IL-12, granulocyte-macrophage colony stimulating factor, tumor necrosis factor-α, interferon-α, interferon-γ], antibodies (αPD-1, αPD-L1, αTIM-3, αOX40, αCTLA-4, αLAG-3), dendritic cell-based vaccines, and chemokines (CXCR2). There are a substantial number of ongoing clinical trials using two or more of these combination therapies. Preliminary results indicate that these combination therapies are a promising area to focus on for cancer treatments, especially melanoma. The main challenges with the combination of cellular and immunotherapies are adverse events due to toxicities and autoimmunity. Identifying mechanisms for reducing or eliminating these adverse events remains a critical area of research. Many important questions still need to be elucidated in regard to combination cellular therapies and immunotherapies, but with the number of ongoing clinical trials, the future of curative melanoma therapies is promising.
Collapse
|
152
|
Abstract
The recent development of effective immune checkpoint inhibition (ICI), first demonstrated in melanoma, has revolutionized cancer treatment. Monoclonal antibodies blocking the immune checkpoints cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 receptor (PD-1) have shown substantial clinical benefit in a subset of patients across tumor types and in both the metastatic and adjuvant settings. In this article, we review the interaction between the immune system and solid tumors, and describe modes of immune response failure and the physiologic role of immune checkpoints. We also review the known mechanisms of immune checkpoint inhibitors, focusing on US FDA-approved agents targeting CTLA-4 and PD-1. Within this framework, we classify hypothesized tumor intrinsic and extrinsic predictive markers for response and resistance to ICI, and map them to their putative underlying biological mechanism. Finally, we outline future directions in ICI, including the development of new therapeutic targets, rational combination therapies, integrated predictive models for individual patients to optimize therapy, and expansion into different disease types.
Collapse
Affiliation(s)
- David Liu
- Division of Medical Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Yawkey 7E, Boston, MA, 02114, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Russell W Jenkins
- Division of Medical Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Yawkey 7E, Boston, MA, 02114, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Ryan J Sullivan
- Division of Medical Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Yawkey 7E, Boston, MA, 02114, USA.
| |
Collapse
|
153
|
Vermaelen K. Vaccine Strategies to Improve Anti-cancer Cellular Immune Responses. Front Immunol 2019; 10:8. [PMID: 30723469 PMCID: PMC6349827 DOI: 10.3389/fimmu.2019.00008] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 01/03/2019] [Indexed: 12/24/2022] Open
Abstract
More than many other fields in medicine, cancer vaccine development has been plagued by a wide gap between the massive amounts of highly encouraging preclinical data on one hand, and the disappointing clinical results on the other. It is clear now that traditional approaches from the infectious diseases' vaccine field cannot be borrowed as such to treat cancer. This review highlights some of the strategies developed to improve vaccine formulations for oncology, including research into more powerful or “smarter” adjuvants to elicit anti-tumoral cellular immune responses. As an illustration of the difficulties in translating smart preclinical strategies into real benefit for the cancer patient, the difficult road of vaccine development in lung cancer is given as example. Finally, an outline is provided of the combinatorial strategies that leverage the increasing knowledge on tumor-associated immune suppressive networks. Indeed, combining with drugs that target the dominant immunosuppressive pathway in a given tumor promises to unlock the true power of cancer vaccines and potentially offer long-term protection from disease relapse.
Collapse
Affiliation(s)
- Karim Vermaelen
- Tumor Immunology Laboratory, Department of Pulmonary Medicine and Immuno-Oncology Network Ghent, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
154
|
Bahig H, Aubin F, Stagg J, Gologan O, Ballivy O, Bissada E, Nguyen-Tan FP, Soulières D, Guertin L, Filion E, Christopoulos A, Lambert L, Tehfe M, Ayad T, Charpentier D, Jamal R, Wong P. Phase I/II trial of Durvalumab plus Tremelimumab and stereotactic body radiotherapy for metastatic head and neck carcinoma. BMC Cancer 2019; 19:68. [PMID: 30642290 PMCID: PMC6332607 DOI: 10.1186/s12885-019-5266-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 01/02/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The efficacy of immunotherapy targeting the PD-1/PD-L1 pathway has previously been demonstrated in metastatic head and neck squamous cell carcinoma (HNSCC). Stereotactic Body Radiotherapy (SBRT) aims at ablating metastatic lesions and may play a synergistic role with immunotherapy. The purpose of this study is to assess the safety and efficacy of triple treatment combination (TTC) consisting of the administration of durvalumab and tremelimumab in combination with SBRT in metastatic HNSCC. METHOD This is a phase I/II single arm study that will include 35 patients with 2-10 extracranial metastatic lesions. Patients will receive durvalumab (1500 mg IV every 4 weeks (Q4W)) and tremelimumab (75 mg IV Q4W for a total of 4 doses) until progression, unacceptable toxicity or patient withdrawal. SBRT to 2-5 metastases will be administered between cycles 2 and 3 of immunotherapy. The safety of the treatment combination will be evaluated through assessment of TTC-related toxicities, defined as grade 3-5 toxicities based on Common Terminology Criteria for Adverse Events (v 4.03), occurring within 6 weeks from SBRT start, and that are definitely, probably or possibly related to the combination of all treatments. We hypothesize that dual targeting of PD-L1 and CTLA-4 pathways combined with SBRT will lead to < 35% grade 3-5 acute toxicities related to TTC. Progression free survival (PFS) will be the primary endpoint of the phase II portion of this study and will be assessed with radiological exams every 8 weeks using the RECIST version 1.1 criteria. DISCUSSION The combination of synergistic dual checkpoints inhibition along with ablative radiation may significantly potentiate the local and systemic disease control. This study constitutes the first clinical trial combining effects of SBRT with dual checkpoint blockade with durvalumab and tremelimumab in the treatment of metastatic HNSCC. If positive, this study would lead to a phase III trial testing this treatment combination against standard of care in metastatic HNSCC. TRIAL REGISTRATION NCT03283605 . Registration date: September 14, 2017; version 1.
Collapse
Affiliation(s)
- Houda Bahig
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Francine Aubin
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Olguta Gologan
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Pathology Department, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Olivier Ballivy
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Eric Bissada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Department of Otorhinolaryngology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Felix-Phuc Nguyen-Tan
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Denis Soulières
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Louis Guertin
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Department of Otorhinolaryngology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Edith Filion
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Apostolos Christopoulos
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Department of Otorhinolaryngology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Louise Lambert
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Mustapha Tehfe
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Tareck Ayad
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
- Department of Otorhinolaryngology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - Danielle Charpentier
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Rahima Jamal
- Department of Medical Oncology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Philip Wong
- Department of Radiation Oncology, Centre Hospitalier de l’Université de Montréal, 1051 Sanguinet Street, Montreal, QC H2X 3E4 Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC Canada
| |
Collapse
|
155
|
Kazaana A, Sano E, Yoshimura S, Makita K, Hara H, Yoshino A, Ueda T. Promotion of TRAIL/Apo2L-induced apoptosis by low-dose interferon-β in human malignant melanoma cells. J Cell Physiol 2019; 234:13510-13524. [PMID: 30613977 DOI: 10.1002/jcp.28029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/30/2018] [Indexed: 12/29/2022]
Abstract
Interferon β (IFN-β) is considered a signaling molecule with important therapeutic potential in cancer since IFN-β-induced gene transcription mediates antiproliferation and cell death induction. Whereas, TNF-related apoptosis inducing ligand/Apo2 ligand (TRAIL/Apo2L) has emerged as a promising anticancer agent because it induces apoptosis specifically in cancer cells. In this study, we elucidated that IFN-β augments TRAIL-induced apoptosis synergistically using five human malignant melanoma cells. All of these cells were induced apoptosis by TRAIL. Whereas, the response against IFN-β was different in amelanotic cells (A375 and CRL1579) and melanotic cells (G361, SK-MEL-28, and MeWo). The responsibility of amelanotic cells against IFN-β was higher than those of melanotic cells. The synergism of IFN-β and TRAIL were correlated with the responsibilities of the cells against IFN-β. The synergistic interaction was confirmed by a combination index based on the Chou-Talalay method. The upregulation of apoptosis in amelanotic cells was caused by very low doses of IFN-β (over 0.1 IU/ml). Both of p53-mediated intrinsic pathway and Fas-related extrinsic pathway were activated by IFN-β alone and combination with TRAIL. Further, TRAIL death receptors (DR4 and DR5) were upregulated by a low-dose IFN-β (over 0.1 IU/ml) and the expression was more promoted by the combination with TRAIL. It was clarified that the upregulation of DR5 is associated with the declination of viability.
Collapse
Affiliation(s)
- Akira Kazaana
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Japan
| | - Emiko Sano
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Japan
| | - Sodai Yoshimura
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Kotaro Makita
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Hiroyuki Hara
- Department of Functional Morphology, Nihon University School of Medicine, Tokyo, Japan
| | - Atsuo Yoshino
- Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Takuya Ueda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Japan
| |
Collapse
|
156
|
Abstract
This chapter focuses on the discovery of the Major Histocompatibility Complex (MHC) in mice (H-2) and in humans (HLA), and on the role played by the International HLA Workshops in the analysis and characterization of this complex genetic system. The early days of Tumour Immunology and the importance of the definition of Tumour Associated Transplantation Antigens (TATA) are also discussed. Today we know that tumour cells can be killed by T lymphocytes by recognizing tumour antigenic peptides presented by MHC molecules and they can also escape this recognition by losing the expression of MHC molecules. This important phenomenon has been profoundly studied for many years both in my lab in Granada and in other laboratories. The results of this research have important implications for the new generation of cancer immunotherapy that boosts T cell responses. A historical perspective of major discoveries is presented in this chapter, with the names of the scientists that have made a significant contribution to the enormous progress made in the field of Tumour Immunology.
Collapse
Affiliation(s)
- Federico Garrido
- Departamento de Analisis Clinicos e Inmunologia, Hospital Universitario Virgen de las Nieves, Facultad de Medicina, Universidad de Granada, Granada, Spain
| |
Collapse
|
157
|
Uddin I, Joshi K, Oakes T, Heather JM, Swanton C, Chain B. An Economical, Quantitative, and Robust Protocol for High-Throughput T Cell Receptor Sequencing from Tumor or Blood. Methods Mol Biol 2019; 1884:15-42. [PMID: 30465193 DOI: 10.1007/978-1-4939-8885-3_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The T cell receptor repertoire provides a window to the cellular adaptive immune response within a tumor, and has the potential to identify specific and personalized biomarkers for tracking host responses during cancer therapy, including immunotherapy. We describe a protocol for amplifying, sequencing, and analyzing T cell receptors which is economical, robust, sensitive, and versatile. The key experimental step is the ligation of a single-stranded oligonucleotide to the 3' end of the T cell receptor cDNA, which allows easy amplification of all possible rearrangements using only a single set of primers per locus, while simultaneously introducing a unique molecular identifier to label each starting cDNA molecule. After sequencing, this molecular identifier can be used to correct both sequence errors and the effects of differential PCR amplification efficiency, thus producing a more accurate measure of the true T cell receptor frequency within the sample. This method has been applied to the analysis of unfractionated human tumor lysates, subpopulations of tumor-infiltrating lymphocytes, and peripheral blood samples from patients with a variety of solid tumors.
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/therapeutic use
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- High-Throughput Nucleotide Sequencing/economics
- High-Throughput Nucleotide Sequencing/instrumentation
- High-Throughput Nucleotide Sequencing/methods
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Neoplasms/blood
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/pathology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Imran Uddin
- Division of Infection and Immunity, UCL, London, UK
| | - Kroopa Joshi
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK
| | - Theres Oakes
- Division of Infection and Immunity, UCL, London, UK
| | | | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, UCL, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Benny Chain
- Division of Infection and Immunity, UCL, London, UK.
| |
Collapse
|
158
|
Wolf Y, Samuels Y. Cancer research in the era of immunogenomics. ESMO Open 2018; 3:e000475. [PMID: 30622743 PMCID: PMC6307593 DOI: 10.1136/esmoopen-2018-000475] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/14/2022] Open
Abstract
The most meaningful advancement in cancer treatment in recent years has been the emergence of immunotherapy. Checkpoint inhibitor blockade and adoptive T cell therapy have shown remarkable clinical effects in a wide range of tumour types. Despite these advances, many tumours do not respond to these treatments, which raises the need to further investigate how patients can benefit from immunotherapy. This effort can now take advantage of the recent technological progress in single-cell, high-throughput sequencing and computational efforts. In this review, we will discuss advances in different immunotherapies and the principles of cancer immunogenomics, with an emphasis on the detection of cancer neoantigens with human leucocyte antigen peptidomics, and how these principles can be further used for more efficient clinical output.
Collapse
Affiliation(s)
- Yochai Wolf
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
159
|
Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat Med 2018; 25:89-94. [PMID: 30510250 DOI: 10.1038/s41591-018-0266-5] [Citation(s) in RCA: 411] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/18/2018] [Indexed: 12/21/2022]
Abstract
Infiltration of human cancers by T cells is generally interpreted as a sign of immune recognition, and there is a growing effort to reactivate dysfunctional T cells at such tumor sites1. However, these efforts only have value if the intratumoral T cell receptor (TCR) repertoire of such cells is intrinsically tumor reactive, and this has not been established in an unbiased manner for most human cancers. To address this issue, we analyzed the intrinsic tumor reactivity of the intratumoral TCR repertoire of CD8+ T cells in ovarian and colorectal cancer-two tumor types for which T cell infiltrates form a positive prognostic marker2,3. Data obtained demonstrate that a capacity to recognize autologous tumor is limited to approximately 10% of intratumoral CD8+ T cells. Furthermore, in two of four patient samples tested, no tumor-reactive TCRs were identified, despite infiltration of their tumors by T cells. These data indicate that the intrinsic capacity of intratumoral T cells to recognize adjacent tumor tissue can be rare and variable, and suggest that clinical efforts to reactivate intratumoral T cells will benefit from approaches that simultaneously increase the quality of the intratumoral TCR repertoire.
Collapse
|
160
|
Maccalli C, Rasul KI, Elawad M, Ferrone S. The role of cancer stem cells in the modulation of anti-tumor immune responses. Semin Cancer Biol 2018; 53:189-200. [DOI: 10.1016/j.semcancer.2018.09.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/12/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023]
|
161
|
de Moel EC, Rozeman EA, Kapiteijn EH, Verdegaal EME, Grummels A, Bakker JA, Huizinga TWJ, Haanen JB, Toes REM, van der Woude D. Autoantibody Development under Treatment with Immune-Checkpoint Inhibitors. Cancer Immunol Res 2018; 7:6-11. [PMID: 30425107 DOI: 10.1158/2326-6066.cir-18-0245] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/03/2018] [Accepted: 11/08/2018] [Indexed: 11/16/2022]
Abstract
Immune-checkpoint inhibitors (ICIs) activate the immune system to assault cancer cells in a manner that is not antigen specific. We hypothesized that tolerance may also be broken to autoantigens, resulting in autoantibody formation, which could be associated with immune-related adverse events (irAEs) and antitumor efficacy. Twenty-three common clinical autoantibodies in pre- and posttreatment sera from 133 ipilimumab-treated melanoma patients were determined, and their development linked to the occurrence of irAEs, best overall response, and survival. Autoantibodies developed in 19.2% (19/99) of patients who were autoantibody-negative pretreatment. A nonsignificant association was observed between development of any autoantibodies and any irAEs [OR, 2.92; 95% confidence interval (CI) 0.85-10.01]. Patients with antithyroid antibodies after ipilimumab had significantly more thyroid dysfunction under subsequent anti-PD-1 therapy: 7/11 (54.6%) patients with antithyroid antibodies after ipilimumab developed thyroid dysfunction under anti-PD1 versus 7/49 (14.3%) patients without antibodies (OR, 9.96; 95% CI, 1.94-51.1). Patients who developed autoantibodies showed a trend for better survival (HR for all-cause death: 0.66; 95% CI, 0.34-1.26) and therapy response (OR, 2.64; 95% CI, 0.85-8.16). We conclude that autoantibodies develop under ipilimumab treatment and could be a potential marker of ICI toxicity and efficacy.
Collapse
Affiliation(s)
- Emma C de Moel
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands.
| | | | - Ellen H Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Els M E Verdegaal
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Annette Grummels
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jaap A Bakker
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - John B Haanen
- Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Diane van der Woude
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
162
|
Hogan SA, Courtier A, Cheng PF, Jaberg-Bentele NF, Goldinger SM, Manuel M, Perez S, Plantier N, Mouret JF, Nguyen-Kim TDL, Raaijmakers MIG, Kvistborg P, Pasqual N, Haanen JBAG, Dummer R, Levesque MP. Peripheral Blood TCR Repertoire Profiling May Facilitate Patient Stratification for Immunotherapy against Melanoma. Cancer Immunol Res 2018; 7:77-85. [PMID: 30425105 DOI: 10.1158/2326-6066.cir-18-0136] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/29/2018] [Accepted: 11/06/2018] [Indexed: 11/16/2022]
Abstract
Many metastatic melanoma patients experience durable responses to anti-PD1 and/or anti-CTLA4; however, a significant proportion (over 50%) do not benefit from the therapies. In this study, we sought to assess pretreatment liquid biopsies for biomarkers that may correlate with response to checkpoint blockade. We measured the combinatorial diversity evenness of the T-cell receptor (TCR) repertoire (the DE50, with low values corresponding to more clonality and lack of TCR diversity) in pretreatment peripheral blood mononuclear cells from melanoma patients treated with anti-CTLA4 (n = 42) or anti-PD1 (n = 38) using a multi-N-plex PCR assay on genomic DNA (gDNA). A receiver operating characteristic curve determined the optimal threshold for a dichotomized analysis according to objective responses as defined by RECIST1.1. Correlations between treatment outcome, clinical variables, and DE50 were assessed in multivariate regression models and confirmed with Fisher exact tests. In samples obtained prior to treatment initiation, we showed that low DE50 values were predictive of a longer progression-free survival and good responses to PD-1 blockade, but, on the other hand, predicted a poor response to CTLA4 inhibition. Multivariate logistic regression models identified DE50 as the only independent predictive factor for response to anti-CTLA4 therapy (P = 0.03) and anti-PD1 therapy (P = 0.001). Fisher exact tests confirmed the association of low DE50 with response in the anti-CTLA4 (P = 0.041) and the anti-PD1 cohort (P = 0.0016). Thus, the evaluation of basal TCR repertoire diversity in peripheral blood, using a PCR-based method, could help predict responses to anti-PD1 and anti-CTLA4 therapies.
Collapse
Affiliation(s)
- Sabrina A Hogan
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Phil F Cheng
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | - Pia Kvistborg
- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | - Reinhard Dummer
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
163
|
Bucktrout SL, Bluestone JA, Ramsdell F. Recent advances in immunotherapies: from infection and autoimmunity, to cancer, and back again. Genome Med 2018; 10:79. [PMID: 30376867 PMCID: PMC6208073 DOI: 10.1186/s13073-018-0588-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
For at least 300 years the immune system has been targeted to improve human health. Decades of work advancing immunotherapies against infection and autoimmunity paved the way for the current explosion in cancer immunotherapies. Pathways targeted for therapeutic intervention in autoimmune diseases can be modulated in the opposite sense in malignancy and infectious disease. We discuss the basic principles of the immune response, how these are co-opted in chronic infection and malignancy, and how these can be harnessed to treat disease. T cells are at the center of immunotherapy. We consider the complexity of T cell functional subsets, differentiation states, and extrinsic and intrinsic influences in the design, success, and lessons from immunotherapies. The integral role of checkpoints in the immune response is highlighted by the rapid advances in FDA approvals and the use of therapeutics that target the CTLA-4 and PD-1/PD-L1 pathways. We discuss the distinct and overlapping mechanisms of CTLA-4 and PD-1 and how these can be translated to combination immunotherapy treatments. Finally, we discuss how the successes and challenges in cancer immunotherapies, such as the collateral damage of immune-related adverse events following checkpoint inhibition, are informing treatment of autoimmunity, infection, and malignancy.
Collapse
Affiliation(s)
- Samantha L Bucktrout
- Parker Institute of Cancer Immunotherapy, 1 Letterman Drive, San Francisco, CA, USA.
| | - Jeffrey A Bluestone
- Parker Institute of Cancer Immunotherapy, 1 Letterman Drive, San Francisco, CA, USA.,Diabetes Center, University of California, San Francisco, San Francisco, CA, 94129, USA
| | - Fred Ramsdell
- Parker Institute of Cancer Immunotherapy, 1 Letterman Drive, San Francisco, CA, USA.
| |
Collapse
|
164
|
Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat Med 2018; 24:1655-1661. [PMID: 30297911 DOI: 10.1038/s41591-018-0198-0] [Citation(s) in RCA: 612] [Impact Index Per Article: 87.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/06/2018] [Indexed: 12/25/2022]
Abstract
Adjuvant ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) both improve relapse-free survival of stage III melanoma patients1,2. In stage IV disease, the combination of ipilimumab + nivolumab is superior to ipilimumab alone and also appears to be more effective than nivolumab monotherapy3. Preclinical work suggests that neoadjuvant application of checkpoint inhibitors may be superior to adjuvant therapy4. To address this question and to test feasibility, 20 patients with palpable stage III melanoma were 1:1 randomized to receive ipilimumab 3 mg kg-1 and nivolumab 1 mg kg-1, as either four courses after surgery (adjuvant arm) or two courses before surgery and two courses postsurgery (neoadjuvant arm). Neoadjuvant therapy was feasible, with all patients undergoing surgery at the preplanned time point. However in both arms, 9/10 patients experienced one or more grade 3/4 adverse events. Pathological responses were achieved in 7/9 (78%) patients treated in the neoadjuvant arm. None of these patients have relapsed so far (median follow-up, 25.6 months). We found that neoadjuvant ipilimumab + nivolumab expand more tumor-resident T cell clones than adjuvant application. While neoadjuvant therapy appears promising, with the current regimen it induced high toxicity rates; therefore, it needs further investigation to preserve efficacy but reduce toxicity.
Collapse
|
165
|
van de Ven R, Hilton TL, Hu HM, Dubay CJ, Haley D, Paustian C, Puri S, Urba WJ, Curti BD, Aung S, Fox BA. Autophagosome-based strategy to monitor apparent tumor-specific CD8 T cells in patients with prostate cancer. Oncoimmunology 2018; 7:e1466766. [PMID: 30524883 PMCID: PMC6279418 DOI: 10.1080/2162402x.2018.1466766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/23/2018] [Accepted: 04/13/2018] [Indexed: 01/05/2023] Open
Abstract
The immune system plays an essential role in eradicating cancer in concert with various treatment modalities. In the absence of autologous tumor material, no standardized method exists to assess T cell responses against the many antigens that may serve as cancer rejection antigens. Thus, development of methods to screen for therapy-induced anti-tumor responses is a high priority that could help tailor therapy. Here we tested whether a tumor-derived antigen source called DRibbles®, which contain a pool of defective ribosomal products (DRiPs), long-lived and short-lived proteins (SLiPs) and danger-associated molecular patterns (DAMPs), can be used to identify tumor-associated antigen (TAA)-specific responses in patients before or after immunotherapy treatment. Protein content, gene expression and non-synonymous - single nucleotide variants (ns-SNVs) present in UbiLT3 DRibbles were compared with prostate adenocarcinomas and the prostate GVAX vaccine cell lines (PC3/LNCaP). UbiLT3 DRibbles were found to share proteins, as well as match tumor sequences for ns-SNVs with prostate adenocarcinomas and with the cell lines PC3 and LNCaP. UbiLT3 DRibbles were used to monitor anti-tumor responses in patients vaccinated with allogeneic prostate GVAX. UbiLT3-DRibble-reactive CD8+ T-cell responses were detected in post-vaccine PBMC of 6/12 patients (range 0.85-22% of CD8+ cells) after 1 week in vitro stimulation (p = 0.007 vs. pre-vaccine). In conclusion, a cancer-derived autophagosome-enriched preparation, packaging over 100 proteins over-expressed in prostate cancer into microvesicles containing DAMPs, could be used to identify CD8+ T cells in peripheral blood from patients after prostate GVAX vaccination and may represent a general method to monitor anti-cancer T cell responses following immunotherapy.
Collapse
Affiliation(s)
- Rieneke van de Ven
- Laboratory of Molecular and Tumor Immunology
- Department of Medical Oncology, VU University medical center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Hong-Ming Hu
- Laboratory of Cancer Immunobiology
- UbiVac LLC, Portland, OR
| | | | | | | | - Sachin Puri
- Laboratory of Molecular and Tumor Immunology
| | - Walter J. Urba
- Robert W. Franz Cancer Research Center at the Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR
| | - Brendan D. Curti
- Robert W. Franz Cancer Research Center at the Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR
| | | | - Bernard A. Fox
- Laboratory of Molecular and Tumor Immunology
- UbiVac LLC, Portland, OR
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| |
Collapse
|
166
|
Yan X, Zhang S, Deng Y, Wang P, Hou Q, Xu H. Prognostic Factors for Checkpoint Inhibitor Based Immunotherapy: An Update With New Evidences. Front Pharmacol 2018; 9:1050. [PMID: 30294272 PMCID: PMC6159743 DOI: 10.3389/fphar.2018.01050] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/31/2018] [Indexed: 02/05/2023] Open
Abstract
Checkpoint inhibitor (CPI) based immunotherapy (i.e., anit-CTLA-4/PD-1/PD-L1 antibodies) can effectively prolong overall survival of patients across several cancer types at the advanced stage. However, only part of patients experience objective responses from such treatments, illustrating large individual differences in terms of both efficacy and adverse drug reactions. Through the observation on a series of CPI based clinical trials in independent patient cohorts, associations of multiple clinical and molecular characteristics with CPI response rate have been determined, including microenvironment, genomic alterations of the cancer cells, and even gut microbiota. A broad interest has been drawn to the question whether and how these prognostic factors can be used as biomarkers for optimal usage of CPIs in precision immunotherapy. Therefore, we reviewed the candidate prognostic factors identified by multiple trials and the experimental investigations, especially those reported in the recent 2 years, and described the possibilities and problems of them in routine clinical usage of cancer treatment as biomarkers.
Collapse
Affiliation(s)
- Xinyu Yan
- Department of Laboratory Medicine, Research Center of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shouyue Zhang
- Department of Laboratory Medicine, Research Center of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yun Deng
- Department of Laboratory Medicine, Research Center of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Peiqi Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianqian Hou
- Department of Laboratory Medicine, Research Center of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Heng Xu
- Department of Laboratory Medicine, Research Center of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China.,Precision Medicine Center, State Key Laboratory of Biotherapy and Precision Medicine, Key Laboratory of Sichuan Province, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| |
Collapse
|
167
|
Wei SC, Duffy CR, Allison JP. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov 2018; 8:1069-1086. [PMID: 30115704 DOI: 10.1158/2159-8290.cd-18-0367] [Citation(s) in RCA: 2106] [Impact Index Per Article: 300.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/04/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023]
Abstract
Immune checkpoint blockade is able to induce durable responses across multiple types of cancer, which has enabled the oncology community to begin to envision potentially curative therapeutic approaches. However, the remarkable responses to immunotherapies are currently limited to a minority of patients and indications, highlighting the need for more effective and novel approaches. Indeed, an extraordinary amount of preclinical and clinical investigation is exploring the therapeutic potential of negative and positive costimulatory molecules. Insights into the underlying biological mechanisms and functions of these molecules have, however, lagged significantly behind. Such understanding will be essential for the rational design of next-generation immunotherapies. Here, we review the current state of our understanding of T-cell costimulatory mechanisms and checkpoint blockade, primarily of CTLA4 and PD-1, and highlight conceptual gaps in knowledge.Significance: This review provides an overview of immune checkpoint blockade therapy from a basic biology and immunologic perspective for the cancer research community. Cancer Discov; 8(9); 1069-86. ©2018 AACR.
Collapse
Affiliation(s)
- Spencer C Wei
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Colm R Duffy
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
168
|
Kobold S, Pantelyushin S, Rataj F, Vom Berg J. Rationale for Combining Bispecific T Cell Activating Antibodies With Checkpoint Blockade for Cancer Therapy. Front Oncol 2018; 8:285. [PMID: 30090763 PMCID: PMC6068270 DOI: 10.3389/fonc.2018.00285] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/09/2018] [Indexed: 12/31/2022] Open
Abstract
T cells have been established as core effectors for cancer therapy; this has moved the focus of therapeutic endeavors to effectively enhance or restore T cell tumoricidal activity rather than directly target cancer cells. Both antibodies targeting the checkpoint inhibitory molecules programmed death receptor 1 (PD1), PD-ligand 1 (PD-L1) and cytotoxic lymphocyte activated antigen 4 (CTLA4), as well as bispecific antibodies targeting CD3 and CD19 are now part of the standard of care. In particular, antibodies to checkpoint molecules have gained broad approval in a number of solid tumor indications, such as melanoma or non-small cell lung cancer based on their unparalleled efficacy. In contrast, the efficacy of bispecific antibody-derivatives is much more limited and evidence is emerging that their activity is regulated through diverse checkpoint molecules. In either case, both types of compounds have their limitations and most patients will not benefit from them in the long run. A major aspect under investigation is the lack of baseline antigen-specific T cells in certain patient groups, which is thought to render responses to checkpoint inhibition less likely. On the other hand, bispecific antibodies are also restricted by induced T cell anergy. Based on these considerations, combination of bispecific antibody mediated on-target T cell activation and reversal of anergy bears high promise. Here, we will review current evidence for such combinatorial approaches, as well as ongoing clinical investigations in this area. We will also discuss potential evidence-driven future avenues for testing.
Collapse
Affiliation(s)
- Sebastian Kobold
- Center of Integrated Protein Science Munich and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Munich, Germany
| | | | - Felicitas Rataj
- Center of Integrated Protein Science Munich and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Munich, Germany
| | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
169
|
Immune Profiling of Cancer Patients Treated with Immunotherapy: Advances and Challenges. Biomedicines 2018; 6:biomedicines6030076. [PMID: 30004433 PMCID: PMC6163220 DOI: 10.3390/biomedicines6030076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/23/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022] Open
Abstract
The recent advances in immunotherapy and the availability of novel drugs to target the tumor microenvironment have dramatically changed the paradigm of cancer treatment. Nevertheless, a significant proportion of cancer patients are unresponsive or develop resistance to these treatments. With the aim to increase the clinical efficacy of immunotherapy, combinations of agents and standard therapies with complementary actions have been developed mostly on an empirical base, since their mechanisms of actions are not yet fully dissected. The characterization of immune responsiveness and its monitoring along with the treatment of cancer patients with immunotherapy can provide insights into the mechanisms of action of these therapeutic regimens and contribute to the optimization of patients’ stratification and of combination strategies and to the prediction of treatment-related toxicities. Thus far, none of the immunomonitoring strategies has been validated for routine clinical practice. Moreover, it is becoming clear that the genomic and molecular make-up of tumors and of the infiltrating immune system represent important determinants of the clinical responses to immunotherapy. This review provides an overview of different approaches for the immune profiling of cancer patients and discusses their advantages and limitations. Recent advances in genomic-based assays and in the identification of host genomic relationships with immune responses represent promising approaches to identify molecular determinants and biomarkers to improve the clinical efficacy of cancer immunotherapy.
Collapse
|
170
|
Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol 2018; 62:29-39. [PMID: 29990692 DOI: 10.1016/j.intimp.2018.06.001] [Citation(s) in RCA: 837] [Impact Index Per Article: 119.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022]
Abstract
Although T lymphocytes have long been appreciated for their role in the immunosurveillance of cancer, it has been the realization that cancer cells may ultimately escape a response from tumor-reactive T cells that has ignited efforts to enhance the efficacy of anti-tumor immune responses. Recent advances in our understanding of T cell immunobiology have been particularly instrumental in informing therapeutic strategies to overcome mechanisms of tumor immune escape, and immune checkpoint blockade has emerged as one of the most promising therapeutic options for patients in the history of cancer treatment. Designed to interfere with inhibitory pathways that naturally constrain T cell reactivity, immune checkpoint blockade releases inherent limits on the activation and maintenance of T cell effector function. In the context of cancer, where negative T cell regulatory pathways are often overactive, immune checkpoint blockade has proven to be an effective strategy for enhancing the effector activity and clinical impact of anti-tumor T cells. Checkpoint inhibitors targeting CTLA-4, PD-1, and PD-L1 have yielded unprecedented and durable responses in a significant percentage of cancer patients in recent years, leading to U.S. FDA approval of six checkpoint inhibitors for numerous cancer indications since 2011. In this review, we highlight the clinical success of these FDA-approved immune checkpoint inhibitors and discuss current challenges and future strategies that must be considered going forward to maximize the efficacy of immune checkpoint blockade therapy for cancer.
Collapse
|
171
|
Pilla L, Ferrone S, Maccalli C. Methods for improving the immunogenicity and efficacy of cancer vaccines. Expert Opin Biol Ther 2018; 18:765-784. [PMID: 29874943 PMCID: PMC8670419 DOI: 10.1080/14712598.2018.1485649] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Cancer vaccines represent one of the oldest immunotherapy strategies. A variety of tumor-associated antigens have been exploited to investigate their immunogenicity as well as multiple strategies for vaccine administration. These efforts have led to the development of several clinical trials in tumors with different histological origins to test the clinical efficacy of cancer vaccines. However, suboptimal clinical results have been reported mainly due to the lack of optimized strategies to induce strong and sustained systemic tumor antigen-specific immune responses. AREAS COVERED We provide an overview of different types of cancer vaccines that have been developed and used in the context of clinical studies. Moreover, we review different preclinical and clinical strategies pursued to enhance the immunogenicity, stability, and targeting at tumor site of cancer vaccines. EXPERT OPINION Additional and appropriate preclinical studies are warranted to optimize the immunogenicity and delivery of cancer vaccines. The appropriate choice of target antigens is challenging; however, the exploitation of neoantigens generated from somatic mutations of tumor cells represents a promising approach to target highly immunogenic tumor-specific antigens. Remarkably, the investigation of the combination of cancer vaccines with immunomodulating agents able to skew the tumor microenvironment from immunosuppressive to immunostimulating will dramatically improve their clinical efficacy.
Collapse
Affiliation(s)
- Lorenzo Pilla
- Medical Oncology Unit, San Gerardo Hospital, Monza, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Maccalli
- Clinical Research Center, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|
172
|
Buder-Bakhaya K, Hassel JC. Biomarkers for Clinical Benefit of Immune Checkpoint Inhibitor Treatment-A Review From the Melanoma Perspective and Beyond. Front Immunol 2018; 9:1474. [PMID: 30002656 PMCID: PMC6031714 DOI: 10.3389/fimmu.2018.01474] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/13/2018] [Indexed: 12/26/2022] Open
Abstract
Background Immune checkpoint inhibition (ICI) with anti-CTLA-4 and/or anti-PD-1 antibodies is standard treatment for metastatic melanoma. Anti-PD-1 (pembrolizumab, nivolumab) and anti-PD-L1 antibodies (atezolizumab, durvalumab, and avelumab) have been approved for treatment of several other advanced malignancies, including non-small-cell lung cancer (NSCLC); renal cell, and urothelial carcinoma; head and neck cancer; gastric, hepatocellular, and Merkel-cell carcinoma; and classical Hodgkin lymphoma. In some of these malignancies approval was based on the detection of biomarkers such as PD-L1 expression or high microsatellite instability. Methods We review the current status of prognostic and predictive biomarkers used in ICI for melanoma and other malignancies. We include clinical, tissue, blood, and stool biomarkers, as well as imaging biomarkers. Results Several biomarkers have been studied in ICI for metastatic melanoma. In clinical practice, pre-treatment tumor burden measured by means of imaging and serum lactate dehydrogenase level is already being used to estimate the likelihood of effective ICI treatment. In peripheral blood, the number of different immune cell types, such as lymphocytes, neutrophils, and eosinophils, as well as different soluble factors, have been correlated with clinical outcome. For intra-tumoral biomarkers, expression of the PD-1 ligand PD-L1 has been found to be of some predictive value for anti-PD-1-directed therapy for NSCLC and melanoma. A high mutational load, particularly when accompanied by neoantigens, seems to facilitate immune response and correlates with patient survival for all entities treated by use of ICI. Tumor microenvironment also seems to be of major importance. Interestingly, even the gut microbiome has been found to correlate with response to ICI, most likely through immuno-stimulatory effects of distinct bacteria. New imaging biomarkers, e.g., for PET, and magnetic resonance imaging are also being investigated, and results suggest they will make early prediction of patient response possible. Conclusion Several promising results are available regarding possible biomarkers for response to ICI, which need to be validated in large clinical trials. A better understanding of how ICI works will enable the development of biomarkers that can predict the response of individual patients.
Collapse
Affiliation(s)
- Kristina Buder-Bakhaya
- Section of Dermatooncology, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Section of Dermatooncology, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
173
|
Li AW, Sobral MC, Badrinath S, Choi Y, Graveline A, Stafford AG, Weaver JC, Dellacherie MO, Shih TY, Ali OA, Kim J, Wucherpfennig KW, Mooney DJ. A facile approach to enhance antigen response for personalized cancer vaccination. NATURE MATERIALS 2018; 17:528-534. [PMID: 29507416 PMCID: PMC5970019 DOI: 10.1038/s41563-018-0028-2] [Citation(s) in RCA: 293] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 01/19/2018] [Indexed: 05/16/2023]
Abstract
Existing strategies to enhance peptide immunogenicity for cancer vaccination generally require direct peptide alteration, which, beyond practical issues, may impact peptide presentation and result in vaccine variability. Here, we report a simple adsorption approach using polyethyleneimine (PEI) in a mesoporous silica microrod (MSR) vaccine to enhance antigen immunogenicity. The MSR-PEI vaccine significantly enhanced host dendritic cell activation and T-cell response over the existing MSR vaccine and bolus vaccine formulations. Impressively, a single injection of the MSR-PEI vaccine using an E7 peptide completely eradicated large, established TC-1 tumours in about 80% of mice and generated immunological memory. When immunized with a pool of B16F10 or CT26 neoantigens, the MSR-PEI vaccine eradicated established lung metastases, controlled tumour growth and synergized with anti-CTLA4 therapy. Our findings from three independent tumour models suggest that the MSR-PEI vaccine approach may serve as a facile and powerful multi-antigen platform to enable robust personalized cancer vaccination.
Collapse
Affiliation(s)
- Aileen Weiwei Li
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Miguel C Sobral
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Soumya Badrinath
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Youngjin Choi
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Amanda Graveline
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Alexander G Stafford
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - James C Weaver
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Maxence O Dellacherie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Ting-Yu Shih
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Omar A Ali
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Suwon, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
174
|
Advanced Melanoma: Current Treatment Options, Biomarkers, and Future Perspectives. Am J Clin Dermatol 2018; 19:303-317. [PMID: 29164492 DOI: 10.1007/s40257-017-0325-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Malignant melanoma accounts for the highest number of deaths from skin cancer, and the prognosis of patients with stage IV disease has historically been poor. Novel insights into both mutations driving tumorigenesis and immune escape mechanisms of these tumors have led to effective treatment options that have revolutionized the treatment of this disease. Targeting the MAPK kinase pathway (with BRAF and MEK inhibitors), as well as targeting checkpoints, such as cytotoxic T-lymphocyte associated protein 4 (CTLA-4) or programmed death 1 (PD-1), have improved overall survival in patients with late-stage melanoma, and biomarker research for personalized therapy is ongoing for each of these treatment modalities. In this review, we will discuss current first-line treatment options, discuss biomarkers supporting treatment decisions, and give an outlook on (combination) therapies we expect to become relevant in the near future.
Collapse
|
175
|
Qiao M, Jiang T, Zhou C. Shining light on advanced NSCLC in 2017: combining immune checkpoint inhibitors. J Thorac Dis 2018; 10:S1534-S1546. [PMID: 29951304 PMCID: PMC5994489 DOI: 10.21037/jtd.2018.04.99] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/11/2018] [Indexed: 12/25/2022]
Abstract
The treatment landscape has changed since the immune checkpoint inhibitors were approved in the treatment of non-small cell lung cancer (NSCLC). Although the promising clinical benefit from programmed death-1/programmed death ligand-1 (PD-1/PD-L1) inhibitors was observed in the second or subsequent line treatment of patients who progressed on chemotherapy, it has a long way for single PD-1/PD-L1 inhibitor to move forward to the frontline without a predictive biomarker. Tumor response is far from satisfactory without selection and primary or acquired resistance to PD-1/PD-L1 inhibitors hampered their utility. Therefore, it is crucial to determine a strategy that can optimize the application of immune checkpoint inhibitors and increase the numbers of the responders. Multiple combination approaches based on PD-1/PD-L1 inhibitors are designed and aimed to boost anti-tumor response and benefit a broader population. In this review, we will integrate the updated clinical data to highlight the four most promising combination strategies in advance NSCLC: combination of checkpoint inhibition with chemotherapy, anti-angiogenesis, immunotherapy and radiotherapy. We further discuss the issues needed to be addressed and perspectives in the context of "combination era".
Collapse
Affiliation(s)
- Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
176
|
Ahrends T, Borst J. The opposing roles of CD4 + T cells in anti-tumour immunity. Immunology 2018; 154:582-592. [PMID: 29700809 PMCID: PMC6050207 DOI: 10.1111/imm.12941] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy focuses mainly on anti-tumour activity of CD8+ cytotoxic T lymphocytes (CTLs). CTLs can directly kill all tumour cell types, provided they carry recognizable antigens. However, CD4+ T cells also play important roles in anti-tumour immunity. CD4+ T cells can either suppress or promote the anti-tumour CTL response, either in secondary lymphoid organs or in the tumour. In this review, we highlight opposing mechanisms of conventional and regulatory T cells at both sites. We outline how current cancer immunotherapy strategies affect both subsets and how selective modulation of each subset is important to maximize the clinical response of cancer patients.
Collapse
Affiliation(s)
- Tomasz Ahrends
- Division of Tumour Biology and ImmunologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jannie Borst
- Division of Tumour Biology and ImmunologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
177
|
Abstract
Therapeutic reinvigoration of tumor-specific T cells has greatly improved clinical outcome in cancer. Nevertheless, many patients still do not achieve durable benefit. Recent evidence from studies in murine and human cancer suggest that intratumoral T cells display a broad spectrum of (dys-)functional states, shaped by the multifaceted suppressive signals that occur within the tumor microenvironment. Here we discuss the current understanding of T cell dysfunction in cancer, the value of novel technologies to dissect such dysfunction at the single cell level, and how our emerging understanding of T cell dysfunction may be utilized to develop personalized strategies to restore antitumor immunity.
Collapse
Affiliation(s)
- Daniela S Thommen
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Ton N Schumacher
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
178
|
Toxicological and pharmacological assessment of AGEN1884, a novel human IgG1 anti-CTLA-4 antibody. PLoS One 2018; 13:e0191926. [PMID: 29617360 PMCID: PMC5884502 DOI: 10.1371/journal.pone.0191926] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/20/2017] [Indexed: 12/15/2022] Open
Abstract
CTLA-4 and CD28 exemplify a co-inhibitory and co-stimulatory signaling axis that dynamically sculpts the interaction of antigen-specific T cells with antigen-presenting cells. Anti-CTLA-4 antibodies enhance tumor-specific immunity through a variety of mechanisms including: blockade of CD80 or CD86 binding to CTLA-4, repressing regulatory T cell function and selective elimination of intratumoral regulatory T cells via an Fcγ receptor-dependent mechanism. AGEN1884 is a novel IgG1 antibody targeting CTLA-4. It potently enhanced antigen-specific T cell responsiveness that could be potentiated in combination with other immunomodulatory antibodies. AGEN1884 was well-tolerated in non-human primates and enhanced vaccine-mediated antigen-specific immunity. AGEN1884 combined effectively with PD-1 blockade to elicit a T cell proliferative response in the periphery. Interestingly, an IgG2 variant of AGEN1884 revealed distinct functional differences that may have implications for optimal dosing regimens in patients. Taken together, the pharmacological properties of AGEN1884 support its clinical investigation as a single therapeutic and combination agent.
Collapse
|
179
|
Genetic alterations and tumor immune attack in Yo paraneoplastic cerebellar degeneration. Acta Neuropathol 2018; 135:569-579. [PMID: 29299667 DOI: 10.1007/s00401-017-1802-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/22/2017] [Accepted: 12/23/2017] [Indexed: 12/23/2022]
Abstract
Paraneoplastic cerebellar degenerations with anti-Yo antibodies (Yo-PCD) are rare syndromes caused by an auto-immune response against neuronal antigens (Ags) expressed by tumor cells. However, the mechanisms responsible for such immune tolerance breakdown are unknown. We characterized 26 ovarian carcinomas associated with Yo-PCD for their tumor immune contexture and genetic status of the 2 onconeural Yo-Ags, CDR2 and CDR2L. Yo-PCD tumors differed from the 116 control tumors by more abundant T and B cells infiltration occasionally organized in tertiary lymphoid structures harboring CDR2L protein deposits. Immune cells are mainly in the vicinity of apoptotic tumor cells, revealing tumor immune attack. Moreover, contrary to un-selected ovarian carcinomas, 65% of our Yo-PCD tumors presented at least one somatic mutation in Yo-Ags, with a predominance of missense mutations. Recurrent gains of the CDR2L gene with tumor protein overexpression were also present in 59% of Yo-PCD patients. Overall, each Yo-PCD ovarian carcinomas carried at least one genetic alteration of Yo-Ags. These data demonstrate an association between massive infiltration of Yo-PCD tumors by activated immune effector cells and recurrent gains and/or mutations in autoantigen-encoding genes, suggesting that genetic alterations in tumor cells trigger immune tolerance breakdown and initiation of the auto-immune disease.
Collapse
|
180
|
Abstract
Small cell lung cancer (SCLC) is a devastating and aggressive neuroendocrine carcinoma of the lung. It accounts for ~15% of lung cancer mortality and has had no improvement in standard treatment options for nearly 30 years. However, there is now hope for change with new therapies and modalities of therapy. Immunotherapies and checkpoint inhibitors are entering clinical practice, selected targeted therapies show promise, and "smart bomb"-based drug/radioconjugates have led to good response in early clinical trials. Additionally, new research insights into the genetics and tumor heterogeneity of SCLC alongside the availability of new tools such as patient-derived or circulating tumor cell xenografts offer the potential to shine light on this beshadowed cancer.
Collapse
|
181
|
Cook KW, Durrant LG, Brentville VA. Current Strategies to Enhance Anti-Tumour Immunity. Biomedicines 2018; 6:E37. [PMID: 29570634 PMCID: PMC6027499 DOI: 10.3390/biomedicines6020037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 12/15/2022] Open
Abstract
The interaction of the immune system with cancer is complex, but new approaches are resulting in exciting therapeutic benefits. In order to enhance the immune response to cancer, immune therapies seek to either induce high avidity immune responses to tumour specific antigens or to convert the tumour to a more pro-inflammatory microenvironment. Strategies, including vaccination, oncolytic viruses, and adoptive cell transfer all seek to induce anti-tumour immunity. To overcome the suppressive tumour microenvironment checkpoint inhibitors and modulators of regulatory cell populations have been investigated. This review summarizes the recent advances in immune therapies and discusses the importance of combination therapies in the treatment of cancers.
Collapse
Affiliation(s)
- Katherine W Cook
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottinghamshire NG5 1PB, UK.
| | - Lindy G Durrant
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottinghamshire NG5 1PB, UK.
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottinghamshire NG5 1PB, UK.
| | - Victoria A Brentville
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottinghamshire NG5 1PB, UK.
| |
Collapse
|
182
|
Sun C, Mezzadra R, Schumacher TN. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018; 48:434-452. [PMID: 29562194 PMCID: PMC7116507 DOI: 10.1016/j.immuni.2018.03.014] [Citation(s) in RCA: 1522] [Impact Index Per Article: 217.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022]
Abstract
Expression of programmed death-ligand 1 (PD-L1) is frequently observed in human cancers. Binding of PD-L1 to its receptor PD-1 on activated T cells inhibits anti-tumor immunity by counteracting T cell-activating signals. Antibody-based PD-1-PD-L1 inhibitors can induce durable tumor remissions in patients with diverse advanced cancers, and thus expression of PD-L1 on tumor cells and other cells in the tumor microenviroment is of major clinical relevance. Here we review the roles of the PD-1-PD-L1 axis in cancer, focusing on recent findings on the mechanisms that regulate PD-L1 expression at the transcriptional, posttranscriptional, and protein level. We place this knowledge in the context of observations in the clinic and discuss how it may inform the design of more precise and effective cancer immune checkpoint therapies.
Collapse
Affiliation(s)
- Chong Sun
- Division of Molecular Oncology & Immunology, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Riccardo Mezzadra
- Division of Molecular Oncology & Immunology, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Ton N Schumacher
- Division of Molecular Oncology & Immunology, Oncode Institute, the Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
183
|
Aptsiauri N, Ruiz-Cabello F, Garrido F. The transition from HLA-I positive to HLA-I negative primary tumors: the road to escape from T-cell responses. Curr Opin Immunol 2018; 51:123-132. [PMID: 29567511 DOI: 10.1016/j.coi.2018.03.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/29/2018] [Accepted: 03/01/2018] [Indexed: 12/29/2022]
Abstract
MHC/HLA class I loss in cancer is one of the main mechanisms of tumor immune escape from T-cell recognition and destruction. Tumor infiltration by T lymphocytes (TILs) and by other immune cells was first described many years ago, but has never been directly and clearly linked to the destruction of HLA-I positive and selection of HLA-I negative tumor cells. The degree and the pattern of lymphocyte infiltration in a tumor nest may depend on antigenicity and the developmental stages of the tumors. In addition, it is becoming evident that HLA-I expression and tumor infiltration have a direct correlation with tumor tissue reorganization. We observed that at early stages (permissive Phase I) tumors are heterogeneous, with both HLA-I positive and HLA-negative cancer cells, and are infiltrated by TILs and M1 macrophages as a part of an active anti-tumor Th1 response. At later stages (encapsulated Phase II), tumor nests are mostly HLA-I negative with immune cells residing in the peri-tumoral stroma, which forms a granuloma-like encapsulated tissue structure. All these tumor characteristics, including tumor HLA-I expression pattern, have an important clinical prognostic value and should be closely and routinely investigated in different types of cancer by immunologists and by pathologists. In this review we summarize our current viewpoint about the alterations in HLA-I expression in cancer and discuss how, when and why tumor HLA-I losses occur. We also provide evidence for the negative impact of tumor HLA-I loss in current cancer immunotherapies, with the focus on reversible ('soft') and irreversible ('hard') HLA-I defects.
Collapse
Affiliation(s)
- Natalia Aptsiauri
- Instituto de Investigacion Biosanitaria ibs, 18014 Granada, Spain; Departamento de Bioquimica, Biologia Molecular e Inmunologia III, Facultad de Medicina, Universidad de Granada, Spain
| | - Francisco Ruiz-Cabello
- Servicio de Analisis Clinicos e Inmunologia, UGC Laboratorio Clinico, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; Instituto de Investigacion Biosanitaria ibs, 18014 Granada, Spain; Departamento de Bioquimica, Biologia Molecular e Inmunologia III, Facultad de Medicina, Universidad de Granada, Spain
| | - Federico Garrido
- Servicio de Analisis Clinicos e Inmunologia, UGC Laboratorio Clinico, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; Instituto de Investigacion Biosanitaria ibs, 18014 Granada, Spain; Departamento de Bioquimica, Biologia Molecular e Inmunologia III, Facultad de Medicina, Universidad de Granada, Spain.
| |
Collapse
|
184
|
Bjoern J, Lyngaa R, Andersen R, Hölmich LR, Hadrup SR, Donia M, Svane IM. Influence of ipilimumab on expanded tumour derived T cells from patients with metastatic melanoma. Oncotarget 2018; 8:27062-27074. [PMID: 28423678 PMCID: PMC5432318 DOI: 10.18632/oncotarget.16003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 02/20/2017] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Tumour infiltrating lymphocyte (TIL) based adoptive cell therapy (ACT) is a promising treatment for patients with advanced melanoma. Retrospective studies suggested an association between previous treatment with anti-CTLA-4 antibodies and long term survival after subsequent ACT. Thus, we hypothesized that treatment with anti-CTLA-4 antibodies can induce favourable changes to be detected in TILs. RESULTS Expanded T cells from Ipilimumab treated patients had a higher proportion of cells expressing CD27, intracellular CTLA-4, TIM-3 and LAG-3. In addition, broader and more frequent T cell responses against common tumour antigens were detected in patients treated with Ipilimumab as compared to anti-CTLA-4 naïve patients. MATERIALS AND METHODS Expanded TILs were obtained from patients with advanced melanoma who had received Ipilimumab in the previous six months, or had not received any type of anti-CTLA-4 antibody. T cell specificity and expression of phenotypic and exhaustion markers were scrutinized as well as functional properties. CONCLUSIONS Ipilimumab may induce tumor-infiltration of T cells of a more naïve phenotype expressing markers related to activation or exhaustion. Additionally, Ipilimumab may increase the frequency of T cells recognizing common tumour associated antigens.
Collapse
Affiliation(s)
- Jon Bjoern
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Lyngaa
- Section for Immunology and Vaccinology, Technical University of Denmark, Copenhagen, Denmark.,Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Andersen
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | | | - Sine Reker Hadrup
- Section for Immunology and Vaccinology, Technical University of Denmark, Copenhagen, Denmark.,Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Marco Donia
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
185
|
Chandran PA, Laske K, Cazaly A, Rusch E, Schmid-Horch B, Rammensee HG, Ottensmeier CH, Gouttefangeas C. Validation of Immunomonitoring Methods for Application in Clinical Studies: The HLA-Peptide Multimer Staining Assay. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2018; 94:342-353. [PMID: 27363684 DOI: 10.1002/cyto.b.21397] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/27/2016] [Accepted: 06/28/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND Validated assays are essential to generate data with defined specificity, consistency, and reliability. Although the process of validation is required for applying immunoassays in the context of clinical studies, reports on systematic validation of in vitro T cell assays are scarce so far. We recently validated our HLA-peptide multimer staining assay in a systematic manner so as to qualify the method for monitoring antigen-specific T cell responses after immunotherapy. METHODS Parameters of the assay, specificity, precision, linearity, sensitivity, and robustness were assessed systematically. Experiments were designed to address specifically each parameter and are detailed. RESULTS Nonspecific multimer staining was below the acceptance limit of 0.02% multimer(+) CD8(+) cells. The assay showed acceptable precision in all dimensions it was repeated (CV < 10%) and also demonstrated a linear detection (R2 > 0.99) of antigen specific cells. CONCLUSIONS We succeeded in validating the HLA-multimer staining assay in a systematic manner. Additionally, we propose a technical framework and recommendations that can be applied for validating other T cell assessment methods. © 2016 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- P Anoop Chandran
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Karoline Laske
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Angelica Cazaly
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, United Kingdom
| | - Elisa Rusch
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | | | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| | - Christian H Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, United Kingdom
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, and German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tuebingen, Tuebingen, Germany
| |
Collapse
|
186
|
Schnell A, Schmidl C, Herr W, Siska PJ. The Peripheral and Intratumoral Immune Cell Landscape in Cancer Patients: A Proxy for Tumor Biology and a Tool for Outcome Prediction. Biomedicines 2018; 6:E25. [PMID: 29495308 PMCID: PMC5874682 DOI: 10.3390/biomedicines6010025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/18/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023] Open
Abstract
Functional systemic and local immunity is required for effective anti-tumor responses. In addition to an active engagement with cancer cells and tumor stroma, immune cells can be affected and are often found to be dysregulated in cancer patients. The impact of tumors on local and systemic immunity can be assessed using a variety of approaches ranging from low-dimensional analyses that are performed on large patient cohorts to multi-dimensional assays that are technically and logistically challenging and are therefore confined to a limited sample size. Many of these strategies have been established in recent years leading to exciting findings. Not only were analyses of immune cells in tumor patients able to predict the clinical course of the disease and patients' survival, numerous studies also detected changes in the immune landscape that correlated with responses to novel immunotherapies. This review will provide an overview of established and novel tools for assessing immune cells in tumor patients and will discuss exemplary studies that utilized these techniques to predict patient outcomes.
Collapse
Affiliation(s)
- Annette Schnell
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany.
| | - Christian Schmidl
- Regensburg Centre for Interventional Immunology and University Medical Center of Regensburg, 93053 Regensburg, Germany.
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany.
- Regensburg Centre for Interventional Immunology and University Medical Center of Regensburg, 93053 Regensburg, Germany.
| | - Peter J Siska
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
187
|
Anti-programmed cell death protein 1 tolerance and efficacy after ipilimumab immunotherapy: observational study of 39 patients. Melanoma Res 2018; 27:110-115. [PMID: 27926587 DOI: 10.1097/cmr.0000000000000313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In patients with ipilimumab (IPI)-refractory melanoma, the anti-programmed cell death proteins 1 (PD1s) nivolumab (NIV) and pembrolizumab (PEM) are considered to be a new standard of treatment. Few data are available on anti-PD1 safety in patients who develop IPI-related severe adverse events (AEs) (grade≥3). The aim of this study was to compare the anti-PD1 safety and efficacy in patients with previous severe toxicity to IPI versus in those showing moderate and no previous IPI-related AEs. This single institution-based observational study included all patients treated with anti-PD1 (PEM or NIV) and previously treated with IPI for unresectable stage III or IV melanoma. The patients enrolled were classified according to the occurrence of IPI-related AEs: group A: no previous IPI-related AEs; group B: mild to moderate IPI-related AEs; and group C: severe to life-threatening IPI-related AEs. The main outcome measure was safety of the anti-PD1 among the three groups. The secondary endpoints included response parameters. Groups A, B, and C included, respectively, 16, 13, and 10 patients. The incidence of severe anti-PD1-related AEs (grades 3-4) was 12, 23, and 10% in groups A, B, and C, respectively. One-year estimates of survival were 52.2, 73.4, and 66.7% among the patients in groups A, B, and C, respectively. The number of patients was too small to enable a meaningful statistical comparison. We did not observe any difference in anti-PD1 toxicity onset incidence according to the occurrence of previous IPI AEs. These reassuring real-life data should be confirmed in a wider analysis.
Collapse
|
188
|
Brown JS, Sundar R, Lopez J. Combining DNA damaging therapeutics with immunotherapy: more haste, less speed. Br J Cancer 2018; 118:312-324. [PMID: 29123260 PMCID: PMC5808021 DOI: 10.1038/bjc.2017.376] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/04/2017] [Accepted: 09/04/2017] [Indexed: 12/14/2022] Open
Abstract
The idea that chemotherapy can be used in combination with immunotherapy may seem somewhat counterproductive, as it can theoretically eliminate the immune cells needed for antitumour immunity. However, much preclinical work has now demonstrated that in addition to direct cytotoxic effects on cancer cells, a proportion of DNA damaging agents may actually promote immunogenic cell death, alter the inflammatory milieu of the tumour microenvironment and/or stimulate neoantigen production, thereby activating an antitumour immune response. Some notable combinations have now moved forward into the clinic, showing promise in phase I-III trials, whereas others have proven toxic, and challenging to deliver. In this review, we discuss the emerging data of how DNA damaging agents can enhance the immunogenic properties of malignant cells, focussing especially on immunogenic cell death, and the expansion of neoantigen repertoires. We discuss how best to strategically combine DNA damaging therapeutics with immunotherapy, and the challenges of successfully delivering these combination regimens to patients. With an overwhelming number of chemotherapy/immunotherapy combination trials in process, clear hypothesis-driven trials are needed to refine the choice of combinations, and determine the timing and sequencing of agents in order to stimulate antitumour immunological memory and improve maintained durable response rates, with minimal toxicity.
Collapse
Affiliation(s)
- Jessica S Brown
- Royal Marsden NHS Foundation Trust, Downs Road, London SM2 5PT, UK
| | - Raghav Sundar
- Royal Marsden NHS Foundation Trust, Downs Road, London SM2 5PT, UK
- Department of Haematology-Oncology, National University Health System, Singapore
| | - Juanita Lopez
- Royal Marsden NHS Foundation Trust, Downs Road, London SM2 5PT, UK
- The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|
189
|
Fontaine M, Vogel I, Van Eycke YR, Galuppo A, Ajouaou Y, Decaestecker C, Kassiotis G, Moser M, Leo O. Regulatory T cells constrain the TCR repertoire of antigen-stimulated conventional CD4 T cells. EMBO J 2018; 37:398-412. [PMID: 29263148 PMCID: PMC5793804 DOI: 10.15252/embj.201796881] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 11/17/2017] [Accepted: 11/27/2017] [Indexed: 01/22/2023] Open
Abstract
To analyze the potential role of Tregs in controlling the TCR repertoire breadth to a non-self-antigen, a TCRβ transgenic mouse model (EF4.1) expressing a limited, yet polyclonal naïve T-cell repertoire was used. The response of EF4.1 mice to an I-Ab-associated epitope of the F-MuLV envelope protein is dominated by clones expressing a Vα2 gene segment, thus allowing a comprehensive analysis of the TCRα repertoire in a relatively large cohort of mice. Control and Treg-depleted EF4.1 mice were immunized, and the extent of the Vα2-bearing, antigen-specific TCR repertoire was characterized by high-throughput sequencing and spectratyping analysis. In addition to increased clonal expansion and acquisition of effector functions, Treg depletion led to the expression of a more diverse TCR repertoire comprising several private clonotypes rarely observed in control mice or in the pre-immune repertoire. Injection of anti-CD86 antibodies in vivo led to a strong reduction in TCR diversity, suggesting that Tregs may influence TCR repertoire diversity by modulating costimulatory molecule availability. Collectively, these studies illustrate an additional mechanism whereby Tregs control the immune response to non-self-antigens.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- B7-2 Antigen/immunology
- Cells, Cultured
- Friend murine leukemia virus/immunology
- Lymphocyte Depletion
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes, Regulatory/immunology
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Martina Fontaine
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Isabel Vogel
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Yves-Rémi Van Eycke
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratories of Image, Signal processing & Acoustics Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Adrien Galuppo
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Yousra Ajouaou
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Christine Decaestecker
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratories of Image, Signal processing & Acoustics Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, London, UK
- Department of Medicine Faculty of Medicine, Imperial College London London, UK
| | - Muriel Moser
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Oberdan Leo
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| |
Collapse
|
190
|
Durgeau A, Virk Y, Corgnac S, Mami-Chouaib F. Recent Advances in Targeting CD8 T-Cell Immunity for More Effective Cancer Immunotherapy. Front Immunol 2018; 9:14. [PMID: 29403496 PMCID: PMC5786548 DOI: 10.3389/fimmu.2018.00014] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/04/2018] [Indexed: 12/18/2022] Open
Abstract
Recent advances in cancer treatment have emerged from new immunotherapies targeting T-cell inhibitory receptors, including cytotoxic T-lymphocyte associated antigen (CTLA)-4 and programmed cell death (PD)-1. In this context, anti-CTLA-4 and anti-PD-1 monoclonal antibodies have demonstrated survival benefits in numerous cancers, including melanoma and non-small-cell lung carcinoma. PD-1-expressing CD8+ T lymphocytes appear to play a major role in the response to these immune checkpoint inhibitors (ICI). Cytotoxic T lymphocytes (CTL) eliminate malignant cells through recognition by the T-cell receptor (TCR) of specific antigenic peptides presented on the surface of cancer cells by major histocompatibility complex class I/beta-2-microglobulin complexes, and through killing of target cells, mainly by releasing the content of secretory lysosomes containing perforin and granzyme B. T-cell adhesion molecules and, in particular, lymphocyte-function-associated antigen-1 and CD103 integrins, and their cognate ligands, respectively, intercellular adhesion molecule 1 and E-cadherin, on target cells, are involved in strengthening the interaction between CTL and tumor cells. Tumor-specific CTL have been isolated from tumor-infiltrating lymphocytes and peripheral blood lymphocytes (PBL) of patients with varied cancers. TCRβ-chain gene usage indicated that CTL identified in vitro selectively expanded in vivo at the tumor site compared to autologous PBL. Moreover, functional studies indicated that these CTL mediate human leukocyte antigen class I-restricted cytotoxic activity toward autologous tumor cells. Several of them recognize truly tumor-specific antigens encoded by mutated genes, also known as neoantigens, which likely play a key role in antitumor CD8 T-cell immunity. Accordingly, it has been shown that the presence of T lymphocytes directed toward tumor neoantigens is associated with patient response to immunotherapies, including ICI, adoptive cell transfer, and dendritic cell-based vaccines. These tumor-specific mutation-derived antigens open up new perspectives for development of effective second-generation therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Aurélie Durgeau
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,ElyssaMed, Paris Biotech Santé, Paris, France
| | - Yasemin Virk
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
191
|
Scheid AD, Van Keulen VP, Felts SJ, Neier SC, Middha S, Nair AA, Techentin RW, Gilbert BK, Jen J, Neuhauser C, Zhang Y, Pease LR. Gene Expression Signatures Characterized by Longitudinal Stability and Interindividual Variability Delineate Baseline Phenotypic Groups with Distinct Responses to Immune Stimulation. THE JOURNAL OF IMMUNOLOGY 2018; 200:1917-1928. [PMID: 29352003 DOI: 10.4049/jimmunol.1701099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/12/2017] [Indexed: 11/19/2022]
Abstract
Human immunity exhibits remarkable heterogeneity among individuals, which engenders variable responses to immune perturbations in human populations. Population studies reveal that, in addition to interindividual heterogeneity, systemic immune signatures display longitudinal stability within individuals, and these signatures may reliably dictate how given individuals respond to immune perturbations. We hypothesize that analyzing relationships among these signatures at the population level may uncover baseline immune phenotypes that correspond with response outcomes to immune stimuli. To test this, we quantified global gene expression in peripheral blood CD4+ cells from healthy individuals at baseline and following CD3/CD28 stimulation at two time points 1 mo apart. Systemic CD4+ cell baseline and poststimulation molecular immune response signatures (MIRS) were defined by identifying genes expressed at levels that were stable between time points within individuals and differential among individuals in each state. Iterative differential gene expression analyses between all possible phenotypic groupings of at least three individuals using the baseline and stimulated MIRS gene sets revealed shared baseline and response phenotypic groupings, indicating the baseline MIRS contained determinants of immune responsiveness. Furthermore, significant numbers of shared phenotype-defining sets of determinants were identified in baseline data across independent healthy cohorts. Combining the cohorts and repeating the analyses resulted in identification of over 6000 baseline immune phenotypic groups, implying that the MIRS concept may be useful in many immune perturbation contexts. These findings demonstrate that patterns in complex gene expression variability can be used to define immune phenotypes and discover determinants of immune responsiveness.
Collapse
Affiliation(s)
- Adam D Scheid
- Immunology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN 55905
| | - Virginia P Van Keulen
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905
| | - Sara J Felts
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905
| | - Steven C Neier
- Immunology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN 55905
| | - Sumit Middha
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN 55905
| | - Asha A Nair
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN 55905
| | - Robert W Techentin
- Special Purpose Processor Development Group, Mayo Clinic, Rochester, MN 55901
| | - Barry K Gilbert
- Special Purpose Processor Development Group, Mayo Clinic, Rochester, MN 55901
| | - Jin Jen
- Medical Genome Facility Gene Expression Core and Department of Experimental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Claudia Neuhauser
- Informatics Institute, University of Minnesota, Minneapolis, MN 55455
| | - Yuji Zhang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN 55905
| | - Larry R Pease
- Immunology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN 55905; .,Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905
| |
Collapse
|
192
|
Efremova M, Rieder D, Klepsch V, Charoentong P, Finotello F, Hackl H, Hermann-Kleiter N, Löwer M, Baier G, Krogsdam A, Trajanoski Z. Targeting immune checkpoints potentiates immunoediting and changes the dynamics of tumor evolution. Nat Commun 2018; 9:32. [PMID: 29296022 PMCID: PMC5750210 DOI: 10.1038/s41467-017-02424-0] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 11/28/2017] [Indexed: 02/07/2023] Open
Abstract
The cancer immunoediting hypothesis postulates a dual role of the immune system: protecting the host by eliminating tumor cells, and shaping the tumor by editing its genome. Here, we elucidate the impact of evolutionary and immune-related forces on editing the tumor in a mouse model for hypermutated and microsatellite-instable colorectal cancer. Analyses of wild-type and immunodeficient RAG1 knockout mice transplanted with MC38 cells reveal that upregulation of checkpoint molecules and infiltration by Tregs are the major tumor escape mechanisms. Our results show that the effects of immunoediting are weak and that neutral accumulation of mutations dominates. Targeting the PD-1/PD-L1 pathway using immune checkpoint blocker effectively potentiates immunoediting. The immunoediting effects are less pronounced in the CT26 cell line, a non-hypermutated/microsatellite-instable model. Our study demonstrates that neutral evolution is another force that contributes to sculpting the tumor and that checkpoint blockade effectively enforces T-cell-dependent immunoselective pressure. The cancer immunoediting hypothesis assumes the immune system sculpts the cancer genome. Here the authors show, in a mouse model, that neutral evolution outweighs the effects of immunoselection and that immune checkpoint blockade potentiates the immunoediting, switching the system to non-neutral evolution.
Collapse
Affiliation(s)
- Mirjana Efremova
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Rieder
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Victoria Klepsch
- Division of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Pornpimol Charoentong
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Francesca Finotello
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Hackl
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Martin Löwer
- TRON -Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Gottfried Baier
- Division of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Anne Krogsdam
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria.
| | - Zlatko Trajanoski
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
193
|
Joshi K, Chain BM, Peggs KS, Quezada SA. The "Achilles' Heel" of Cancer and Its Implications for the Development of Novel Immunotherapeutic Strategies. Cold Spring Harb Perspect Med 2018; 8:a027086. [PMID: 28630228 PMCID: PMC5749142 DOI: 10.1101/cshperspect.a027086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the last century, scientists have embraced the idea of mobilizing antitumor immune responses in patients with cancer. In the last decade, we have seen the rebirth of cancer immunotherapy and its validation in a series of high profile clinical trials following the discovery of several immune-regulatory receptors. Recent studies point toward the tumor mutational load and resulting neoantigen burden as being crucial to tumor cell recognition by the immune system, highlighting a potentially targetable Achilles' heel in cancer. In this review, we explore the key mechanisms that underpin the recognition of cancerous cells by the immune system and discuss how we may advance immunotherapeutic strategies to target the cancer mutanome to stimulate tumor-specific immune responses, ultimately, to improve the clinical outcome for patients with cancer.
Collapse
Affiliation(s)
- Kroopa Joshi
- Cancer Immunology Unit, University College London Cancer Institute, London WC1E 6BT, United Kingdom
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, United Kingdom
| | - Benjamin M Chain
- Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - Karl S Peggs
- Cancer Immunology Unit, University College London Cancer Institute, London WC1E 6BT, United Kingdom
| | - Sergio A Quezada
- Cancer Immunology Unit, University College London Cancer Institute, London WC1E 6BT, United Kingdom
| |
Collapse
|
194
|
Song M, Chen X, Wang L, Zhang Y. Future of anti-PD-1/PD-L1 applications: Combinations with other therapeutic regimens. Chin J Cancer Res 2018; 30:157-172. [PMID: 29861603 DOI: 10.21147/j.issn.1000-9604.2018.02.01] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Programmed cell death 1 (PD-1)/programmed cell death 1 ligand (PD-L1) blockade has shown promising effects in cancer immunotherapy. Removing the so-called " brakes" on T cell immune responses by blocking the PD-1/PD-L1 check point should boost anti-tumor immunity and provide durable tumor regression for cancer patients. However, 30%-60% of patients show no response to PD-1/PD-L1 blockade. Thus, it is urgent to explore the underlying resistance mechanisms to improve sensitivity to anti-PD-1/PD-L1 therapy. We propose that the mechanisms promoting resistance mainly include T cell exclusion or exhaustion at the tumor site, immunosuppressive factors in the tumor microenvironment (TME), and a range of tumor-intrinsic factors. This review highlights the power of studying the cellular and molecular mechanisms of resistance to improve the rational design of combination therapeutic strategies that can be translated to the clinic. Here, we briefly discuss the development of PD-1/PD-L1 blockade agents and focus on the current issues and future prospects for potential combinatorial therapeutic strategies that include anti-PD-1/PD-L1 therapy, based upon the available preclinical and clinical data.
Collapse
Affiliation(s)
- Mengjia Song
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xinfeng Chen
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Liping Wang
- Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yi Zhang
- Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,School of Life Sciences, Zhengzhou University, Zhengzhou 450052, China.,Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou 450052, China
| |
Collapse
|
195
|
Toulmonde M, Penel N, Adam J, Chevreau C, Blay JY, Le Cesne A, Bompas E, Piperno-Neumann S, Cousin S, Grellety T, Ryckewaert T, Bessede A, Ghiringhelli F, Pulido M, Italiano A. Use of PD-1 Targeting, Macrophage Infiltration, and IDO Pathway Activation in Sarcomas: A Phase 2 Clinical Trial. JAMA Oncol 2018; 4:93-97. [PMID: 28662235 PMCID: PMC5833654 DOI: 10.1001/jamaoncol.2017.1617] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/14/2017] [Indexed: 12/21/2022]
Abstract
IMPORTANCE There is a strong rationale for treating sarcomas with immunotherapy. OBJECTIVE To assess the efficacy and safety of programmed cell death protein 1 (PD-1) targeting in combination with metronomic chemotherapy in sarcomas. DESIGN, SETTING, AND PARTICIPANTS This was an open-label, multicenter, phase 2 study of 4 cohorts of patients with advanced soft-tissue sarcoma (STS), including leiomyosarcoma (LMS), undifferentiated pleomorphic sarcoma (UPS), other sarcomas (others), and gastrointestinal stromal tumor (GIST). All patients received 50 mg twice daily cyclophosphamide 1 week on and 1 week off and 200 mg of intravenous pembrolizumab every 3 weeks. INTERVENTION OR EXPOSURE Pembrolizumab in combination with metronomic cyclophosphamide. MAIN OUTCOMES AND MEASURES There was a dual primary end point, encompassing both the nonprogression and objective responses at 6 months per Response Evaluation Criteria in Solid Tumours (RECIST) v1.1 for LMS, UPS, and others and 6-month nonprogression for GIST. An objective response rate of 20% and/or a 6-month nonprogression rate of 60% were determined as reasonable objectives for treatment with meaningful effect. Correlative studies of immune biomarkers were planned from patient tumor and plasma samples. RESULTS Between June 2015 and July 2016, 57 patients were included (median [range] age, 59.5 [18.5-84.0] years; 24 women [42%]); 50 patients were assessable for the efficacy end point. Three patients experienced tumor shrinkage, resulting in a partial response in a single solitary fibrous tumor. The 6-month nonprogression rates were 0%, 0%, 14.3% (95% CI, 1.8%-42.8%) for LMS, UPS, and others, respectively, and 11.1% (95% CI, 2.8%-48.3%) for GIST. The most frequent adverse events were grade 1 or 2 fatigue, diarrhea, and anemia. The only patient who experienced partial response was the only one with strong programmed cell death 1 ligand 1-positive staining in immune cell. Strong infiltration by macrophage expressing the inhibitory enzyme indoleamine 2,3-dioxygenase 1 (IDO1) was observed in the majority of cases. Moreover, a significant increase in the kynurenine to tryptophan ratio was observed in patient plasma samples during the study treatment. CONCLUSIONS AND RELEVANCE We found that PD-1 inhibition has limited activity in selected STS and GIST. This may be explained by an immunosuppressive tumor microenvironment resulting from macrophage infiltration and IDO1 pathway activation. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT02406781.
Collapse
Affiliation(s)
- Maud Toulmonde
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret, Bordeaux, France
| | - Julien Adam
- Department of Pathology, Gustave Roussy, Villejuif, France
- INSERM U981, Villejuif, France
| | | | - Jean-Yves Blay
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Axel Le Cesne
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| | - Emmanuelle Bompas
- Department of Medical Oncology, Institut de Cancérologie de l’Ouest, Nantes, France
| | | | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Thomas Grellety
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | | | | | | | - Marina Pulido
- Unité de Recherche et d’Epidémiologie Cliniques, Institut Bergonié, Bordeaux, France
- INSERM CIC 1401, Bordeaux, France
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| |
Collapse
|
196
|
Chapuis AG, Roberts IM, Thompson JA, Margolin KA, Bhatia S, Lee SM, Sloan HL, Lai IP, Farrar EA, Wagener F, Shibuya KC, Cao J, Wolchok JD, Greenberg PD, Yee C. T-Cell Therapy Using Interleukin-21-Primed Cytotoxic T-Cell Lymphocytes Combined With Cytotoxic T-Cell Lymphocyte Antigen-4 Blockade Results in Long-Term Cell Persistence and Durable Tumor Regression. J Clin Oncol 2017; 34:3787-3795. [PMID: 27269940 DOI: 10.1200/jco.2015.65.5142] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Peripheral blood-derived antigen-specific cytotoxic T cells (CTLs) provide a readily available source of effector cells that can be administered with minimal toxicity in an outpatient setting. In metastatic melanoma, this approach results in measurable albeit modest clinical responses in patients resistant to conventional therapy. We reasoned that concurrent cytotoxic T-cell lymphocyte antigen-4 (CTLA-4) checkpoint blockade might enhance the antitumor activity of adoptively transferred CTLs. Patients and Methods Autologous MART1-specific CTLs were generated by priming with peptide-pulsed dendritic cells in the presence of interleukin-21 and enriched by peptide-major histocompatibility complex multimer-guided cell sorting. This expeditiously yielded polyclonal CTL lines uniformly expressing markers associated with an enhanced survival potential. In this first-in-human strategy, 10 patients with stage IV melanoma received the MART1-specific CTLs followed by a standard course of anti-CTLA-4 (ipilimumab). Results The toxicity profile of the combined treatment was comparable to that of ipilimumab monotherapy. Evaluation of best responses at 12 weeks yielded two continuous complete remissions, one partial response (PR) using RECIST criteria (two PRs using immune-related response criteria), and three instances of stable disease. Infused CTLs persisted with frequencies up to 2.9% of CD8+ T cells for as long as the patients were monitored (up to 40 weeks). In patients who experienced complete remissions, PRs, or stable disease, the persisting CTLs acquired phenotypic and functional characteristics of long-lived memory cells. Moreover, these patients also developed responses to nontargeted tumor antigens (epitope spreading). Conclusion We demonstrate that combining antigen-specific CTLs with CTLA-4 blockade is safe and produces durable clinical responses, likely reflecting both enhanced activity of transferred cells and improved recruitment of new responses, highlighting the promise of this strategy.
Collapse
Affiliation(s)
- Aude G Chapuis
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ilana M Roberts
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John A Thompson
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kim A Margolin
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Shailender Bhatia
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sylvia M Lee
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Heather L Sloan
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ivy P Lai
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Erik A Farrar
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Felecia Wagener
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kendall C Shibuya
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jianhong Cao
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jedd D Wolchok
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Philip D Greenberg
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Cassian Yee
- Aude G. Chapuis, Ilana M. Roberts, Sylvia M. Lee, Heather L. Sloan, Ivy P. Lai, Erik A. Farrar, Felecia Wagener, Kendall C. Shibuya, Jianhong Cao, Philip D. Greenberg, and Cassian Yee, Fred Hutchinson Cancer Research Center; John A. Thompson, Kim A. Margolin, and Shailender Bhatia, Seattle Cancer Care Alliance and University of Washington, Seattle WA; and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
197
|
Guy TV, Terry AM, Bolton HA, Hancock DG, Zhu E, Brink R, McGuire HM, Shklovskaya E, Fazekas de St. Groth B. Collaboration between tumor-specific CD4+ T cells and B cells in anti-cancer immunity. Oncotarget 2017; 7:30211-29. [PMID: 27121060 PMCID: PMC5058675 DOI: 10.18632/oncotarget.8797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/06/2016] [Indexed: 12/02/2022] Open
Abstract
The role of B cells and antibodies in anti-tumor immunity is controversial, with both positive and negative effects reported in animal models and clinical studies. We developed a murine B16.F10 melanoma model to study the effects of collaboration between tumor-specific CD4+ T cells and B cells on tumor control. By incorporating T cell receptor transgenic T cells and B cell receptor isotype switching B cells, we were able to track the responses of tumor-reactive T and B cells and the development of anti-tumor antibodies in vivo. In the presence of tumor-specific B cells, the number of tumor-reactive CD4+ T cells was reduced in lymphoid tissues and the tumor itself, and this correlated with poor tumor control. B cells had little effect on the Th1 bias of the CD4+ T cell response, and the number of induced FoxP3+ regulatory cells (iTregs) generated from within the original naive CD4+ T cell inoculum was unrelated to the degree of B cell expansion. In response to CD4+ T cell help, B cells produced a range of isotype-switched anti-tumor antibodies, principally IgG1, IgG2a/c and IgG2b. In the absence of CD4+ T cells, B cells responded to agonistic anti-CD40 administration by switching to production of IgG2a/c and, to a lesser extent, IgG1, IgG3, IgA and IgE, which reduced the number of lung metastases after i.v. tumor inoculation but had no effect on the growth of subcutaneous tumors.
Collapse
Affiliation(s)
- Thomas V Guy
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra M Terry
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Holly A Bolton
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - David G Hancock
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Erhua Zhu
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Robert Brink
- B Cell Laboratory, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Helen M McGuire
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Elena Shklovskaya
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Barbara Fazekas de St. Groth
- T Cell Biology Laboratory, Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Sydney, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
198
|
Simon S, Vignard V, Varey E, Parrot T, Knol AC, Khammari A, Gervois N, Lang F, Dreno B, Labarriere N. Emergence of High-Avidity Melan-A–Specific Clonotypes as a Reflection of Anti–PD-1 Clinical Efficacy. Cancer Res 2017; 77:7083-7093. [DOI: 10.1158/0008-5472.can-17-1856] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/29/2017] [Accepted: 09/19/2017] [Indexed: 11/16/2022]
Abstract
Abstract
Therapeutic strategies using anti–PD-1–blocking antibodies reported unparalleled effectiveness for melanoma immunotherapy, but deciphering immune responses modulated by anti–PD-1 treatment remains a crucial issue. Here, we analyzed the composition and functions of the large Melan-A–specific T-cell repertoire in the peripheral blood of 9 melanoma patients before and after 2 months of treatment with anti–PD-1. We observed amplification of Melan-A–specific Vß subfamilies undetectable before therapy (thereafter called emerging Vß subfamilies) in responding patients, with a predominant expansion in patients with a complete response. These emerging Vß subfamilies displayed a higher functional avidity for their cognate antigen than Vß subfamilies not amplified upon anti–PD-1 therapy and could be identified by a sustained coexpression of PD-1 and TIGIT receptors. Thus, in addition to the emergence of neoantigen-specific T cells previously documented upon anti–PD-1 therapy, our work describes the emergence of high-avidity Melan-A–specific clonotypes as a surrogate marker of treatment efficacy. Cancer Res; 77(24); 7083–93. ©2017 AACR.
Collapse
Affiliation(s)
- Sylvain Simon
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
| | - Virginie Vignard
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 3CHU Nantes, Nantes, France
| | - Emilie Varey
- 4Department of Dermato-cancerology of Nantes Hospital, Nantes, France
| | - Tiphaine Parrot
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
| | - Anne-Chantal Knol
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 4Department of Dermato-cancerology of Nantes Hospital, Nantes, France
| | - Amir Khammari
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 4Department of Dermato-cancerology of Nantes Hospital, Nantes, France
| | - Nadine Gervois
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
| | - Francois Lang
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
| | - Brigitte Dreno
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 4Department of Dermato-cancerology of Nantes Hospital, Nantes, France
| | - Nathalie Labarriere
- 1CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
- 2LabEx IGO “Immunotherapy, Graft, Oncology,” Nantes, France
- 3CHU Nantes, Nantes, France
| |
Collapse
|
199
|
Rao M, Zhenjiang L, Meng Q, Sinclair G, Dodoo E, Maeurer M. Mutant Epitopes in Cancer. Oncoimmunology 2017. [DOI: 10.1007/978-3-319-62431-0_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
200
|
Abstract
The identification of predictive biomarkers for the benefit of cancer immunotherapy is the holy grail of the burgeoning immunotherapy field. Recent work has shown that there are a core of concepts that establish the presence of an immune cell-infiltrate, an inflammatory signature of the tumor microenvironment, and the availability of target antigens defined by mutated neoantigens, as critical for the success of the checkpoint blockade. Genetic analyses have shown that resistance to PD-1 blockade, either innate or adaptive, may be due to existing or de novo mutations in signaling pathways critical for T-cell function in a modest proportion of cases. Major hurdles in the field that remain to be overcome are the difficulty of obtaining tumor biopsies for biomarker assessment, the heterogeneity of biomarker expression within tumors and within different tumors from the same patient, and the inducibility of some biomarkers by disease-related processes. Although assessment of peripheral blood or serum biomarkers would be ideal, few data suggest that they would reliably predict outcome with checkpoint blockade. Ultimately, some amalgamated biomarker that includes tumor and host factors will be required to predict which patients are likely to benefit from, or be resistant to, the effects of checkpoint inhibition.
Collapse
Affiliation(s)
- Jeffrey S Weber
- From the Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY
| |
Collapse
|