1
|
Ghaneialvar H, Jahani S, Hashemi E, Khalilzad MA, Falahi S, Rashidi MA, Majidpoor J, Najafi S. Combining anti-checkpoint immunotherapies and cancer vaccines as a novel strategy in oncological therapy: A review. Hum Immunol 2025; 86:111209. [PMID: 39662393 DOI: 10.1016/j.humimm.2024.111209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
The field of cancer immunotherapy has experienced remarkable advancements in the treatment of human cancers over recent decades. Therapeutic cancer vaccines have been employed to elicit antitumor immune responses through the generation of specific reactions against tumor-associated antigens. Although preclinical studies have demonstrated hopeful results and at least one product is approved for clinical use, the overall efficacy of cancer vaccines remains restricted. The co-administration of anti-checkpoint antibodies alongside cancer vaccines is proposed as a potential strategy to enhance the clinical efficacy of immunotherapies. Among the various anti-checkpoint agents, monoclonal antibodies targeting CD127, OX40, and CD40 have been further investigated in combined administration with cancer vaccines, demonstrating a synergistic impact on disease outcomes in both animal models and human subjects. This combinational approach has been shown to suppress tumor regression, improve survival rates, and promote the efficacy of cancer vaccines via multiple mechanisms, including the augmentation of generation, activation, and expansion of CD8+ T cells, as well as the production of tumor-inhibitory cytokines. Importantly, the impact of the concurrent administration of anti-checkpoint agents and cancer vaccines surpass those observed with the sole vaccine, indicating that this strategy may offer significant advantages for clinical application in cancer patients. In this review, we aim to provide a comprehensive overview of the significance and therapeutic potential of the combined administration of checkpoint agonist/antagonist antibodies and cancer vaccines for human tumors.
Collapse
Affiliation(s)
- Hori Ghaneialvar
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Saleheh Jahani
- Department of Pathology, School of Medicine, University of California, San Diego, USA
| | - Elham Hashemi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Shahab Falahi
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Mohammad Amin Rashidi
- Department of Occupational Health and Safety, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran.
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
3
|
Xie Q, Liu X, Liu R, Pan J, Liang J. Cellular mechanisms of combining innate immunity activation with PD-1/PD-L1 blockade in treatment of colorectal cancer. Mol Cancer 2024; 23:252. [PMID: 39529058 PMCID: PMC11555832 DOI: 10.1186/s12943-024-02166-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
PD-1/PD-L1 blockade therapies have displayed extraordinary clinical efficacy for melanoma, renal, bladder and lung cancer; however, only a minority of colorectal cancer (CRC) patients benefit from these treatments. The efficacy of PD-1/PD-L1 blockade in CRC is limited by the complexities of tumor microenvironment. PD-1/PD-L1 blockade immunotherapy is based on T cell-centered view of tumor immunity. However, the onset and maintenance of T cell responses and the development of long-lasting memory T cells depend on innate immune responses. Acknowledging the pivotal role of innate immunity in anti-tumor immune response, this review encapsulates the employment of combinational therapies those involve PD-1/PD-L1 blockade alongside the activation of innate immunity and explores the underlying cellular mechanisms, aiming to harnessing innate immune responses to induce long-lasting tumor control for CRC patients who received PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Xiaolin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Rengyun Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
| |
Collapse
|
4
|
Phoon YP, Lopes JE, Pfannenstiel LW, Marcela Diaz-Montero C, Tian YF, Ernstoff MS, Funchain P, Ko JS, Winquist R, Losey HC, Melenhorst JJ, Gastman BR. Autologous human preclinical modeling of melanoma interpatient clinical responses to immunotherapeutics. J Immunother Cancer 2024; 12:e008066. [PMID: 38604813 PMCID: PMC11015209 DOI: 10.1136/jitc-2023-008066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Despite recent advances in immunotherapy, a substantial population of late-stage melanoma patients still fail to achieve sustained clinical benefit. Lack of translational preclinical models continues to be a major challenge in the field of immunotherapy; thus, more optimized translational models could strongly influence clinical trial development. To address this unmet need, we designed a preclinical model reflecting the heterogeneity in melanoma patients' clinical responses that can be used to evaluate novel immunotherapies and synergistic combinatorial treatment strategies. Using our all-autologous humanized melanoma mouse model, we examined the efficacy of a novel engineered interleukin 2 (IL-2)-based cytokine variant immunotherapy. METHODS To study immune responses and antitumor efficacy for human melanoma tumors, we developed an all-autologous humanized melanoma mouse model using clinically annotated, matched patient tumor cells and peripheral blood mononuclear cells (PBMCs). After inoculating immunodeficient NSG mice with patient tumors and an adoptive cell transfer of autologous PBMCs, mice were treated with anti-PD-1, a novel investigational engineered IL-2-based cytokine (nemvaleukin), or recombinant human IL-2 (rhIL-2). The pharmacodynamic effects and antitumor efficacy of these treatments were then evaluated. We used tumor cells and autologous PBMCs from patients with varying immunotherapy responses to both model the diversity of immunotherapy efficacy observed in the clinical setting and to recapitulate the heterogeneous nature of melanoma. RESULTS Our model exhibited long-term survival of engrafted human PBMCs without developing graft-versus-host disease. Administration of an anti-PD-1 or nemvaleukin elicited antitumor responses in our model that were patient-specific and were found to parallel clinical responsiveness to checkpoint inhibitors. An evaluation of nemvaleukin-treated mice demonstrated increased tumor-infiltrating CD4+ and CD8+ T cells, preferential expansion of non-regulatory T cell subsets in the spleen, and significant delays in tumor growth compared with vehicle-treated controls or mice treated with rhIL-2. CONCLUSIONS Our model reproduces differential effects of immunotherapy in melanoma patients, capturing the inherent heterogeneity in clinical responses. Taken together, these data demonstrate our model's translatability for novel immunotherapies in melanoma patients. The data are also supportive for the continued clinical investigation of nemvaleukin as a novel immunotherapeutic for the treatment of melanoma.
Collapse
Affiliation(s)
- Yee Peng Phoon
- Center for Immunotherapy and Precision Immuno-Oncology (CITI), Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Claudia Marcela Diaz-Montero
- Center for Immunotherapy and Precision Immuno-Oncology (CITI), Cleveland Clinic, Cleveland, Ohio, USA
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ye F Tian
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Pauline Funchain
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | | | - Jan Joseph Melenhorst
- Center for Immunotherapy and Precision Immuno-Oncology (CITI), Cleveland Clinic, Cleveland, Ohio, USA
| | - Brian R Gastman
- Center for Immunotherapy and Precision Immuno-Oncology (CITI), Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
5
|
Dillman RO, Nistor GI, Keirstead HS. Autologous dendritic cells loaded with antigens from self-renewing autologous tumor cells as patient-specific therapeutic cancer vaccines. Hum Vaccin Immunother 2023:2198467. [PMID: 37133853 DOI: 10.1080/21645515.2023.2198467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
A promising personal immunotherapy is autologous dendritic cells (DC) loaded ex vivo with autologous tumor antigens (ATA) derived from self-renewing autologous cancer cells. DC-ATA are suspended in granulocyte-macrophage colony stimulating factor at the time of each subcutaneous injection. Previously, irradiated autologous tumor cell vaccines have produced encouraging results in 150 cancer patients, but the DC-ATA vaccine demonstrated superiority in single-arm and randomized trials in metastatic melanoma. DC-ATA have been injected into more than 200 patients with melanoma, glioblastoma, and ovarian, hepatocellular, and renal cell cancers. Key observations include: [1] greater than 95% success rates for tumor cell cultures and monocyte collection for dendritic cell production; [2] injections are well-tolerated; [3] the immune response is rapid and includes primarily TH1/TH17 cellular responses; [4] efficacy has been suggested by delayed but durable complete tumor regressions in patients with measurable disease, by progression-free survival in glioblastoma, and by overall survival in melanoma.
Collapse
Affiliation(s)
| | - Gabriel I Nistor
- Research and Development, AIVITA Biomedical Inc, Irvine, CA, USA
| | | |
Collapse
|
6
|
Estrada J, Zhan J, Mitchell P, Werner J, Beltran PJ, DeVoss J, Qing J, Cooke KS. OncoVEX mGM-CSFexpands tumor antigen-specific CD8+ T-cell response in preclinical models. J Immunother Cancer 2023; 11:jitc-2022-006374. [PMID: 37164449 PMCID: PMC10173969 DOI: 10.1136/jitc-2022-006374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) have demonstrated clinical efficacy in advanced melanoma, but only a subset of patients with inflamed tumors are responsive. Talimogene laherparepvec (T-VEC), a modified herpes simplex virus type 1 (HSV-1) expressing granulocyte-macrophage colony-stimulating factor (GM-CSF), is a first-in-class oncolytic immunotherapy approved for the treatment of melanoma and has been shown to inflame the tumor microenvironment. To evaluate the potential and mechanisms of T-VEC to elicit systemic antitumor immunity and overcome resistance to checkpoint inhibitors in murine tumor models, OncoVEXmGM-CSF was developed similarly to T-VEC, except the human GM-CSF transgene was replaced with murine GM-CSF. Previous work had demonstrated that OncoVEXmGM-CSF generated systemic antitumor immunity dependent on CD8+ T cells in an immune checkpoint-sensitive tumor cell model. METHODS A novel B16F10 syngeneic tumor model with both HSV-1-permissive subcutaneous tumors and HSV-1-refractory experimental lung metastasis was used to study the local and systemic effects of OncoVEXmGM-CSF treatment alone or in combination with checkpoint inhibitors. RESULTS Intratumoral injection of OncoVEXmGM-CSF in combination with an anti-CTLA-4 or anti-PD-1 blocking antibody led to increased tumor growth inhibition, a reduction in the number of lung metastases, and prolonged animal survival. OncoVEXmGM-CSF induced both neoantigen-specific and tumor antigen-specific T-cell responses. Furthermore, cured mice from the combination treatment of OncoVEXmGM-CSF and anti-CTLA-4 antibody rejected tumor rechallenges. CONCLUSIONS These data support the concept that T-VEC and checkpoint inhibition may be an effective combination to treat patients with advanced melanoma.
Collapse
|
7
|
Ben-Akiva E, Hickey JW, Meyer RA, Isser A, Shannon SR, Livingston NK, Rhodes KR, Kosmides AK, Warren TR, Tzeng SY, Schneck JP, Green JJ. Shape matters: Biodegradable anisotropic nanoparticle artificial antigen presenting cells for cancer immunotherapy. Acta Biomater 2023; 160:187-197. [PMID: 36812956 PMCID: PMC10335041 DOI: 10.1016/j.actbio.2023.02.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
Artificial antigen presenting cells are biomimetic particles that recapitulate the signals presented by natural antigen presenting cells in order to stimulate T cells in an antigen-specific manner using an acellular platform. We have engineered an enhanced nanoscale biodegradable artificial antigen presenting cell by modulating particle shape to achieve a nanoparticle geometry that allows for increased radius of curvature and surface area for T cell contact. The non-spherical nanoparticle artificial antigen presenting cells developed here have reduced nonspecific uptake and improved circulation time compared both to spherical nanoparticles and to traditional microparticle technologies. Additionally, the anisotropic nanoparticle artificial antigen presenting cells efficiently engage with and activate T cells, ultimately leading to a marked anti-tumor effect in a mouse melanoma model that their spherical counterparts were unable to achieve. STATEMENT OF SIGNIFICANCE: Artificial antigen presenting cells (aAPC) can activate antigen-specific CD8+ T cells but have largely been limited to microparticle-based platforms and ex vivo T cell expansion. Although more amenable to in vivo use, nanoscale aAPC have traditionally been ineffective due to limited surface area available for T cell interaction. In this work, we engineered non-spherical biodegradable nanoscale aAPC to investigate the role of particle geometry and develop a translatable platform for T cell activation. The non-spherical aAPC developed here have increased surface area and a flatter surface for T cell engagement and, therefore, can more effectively stimulate antigen-specific T cells, resulting in anti-tumor efficacy in a mouse melanoma model.
Collapse
Affiliation(s)
- Elana Ben-Akiva
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Randall A Meyer
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ariel Isser
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sydney R Shannon
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Natalie K Livingston
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kelly R Rhodes
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Tiarra R Warren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jonathan P Schneck
- Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and the Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
8
|
Chakravarti AR, Groer CE, Gong H, Yudistyra V, Forrest ML, Berkland CJ. Design of a Tumor Binding GMCSF as Intratumoral Immunotherapy of Solid Tumors. Mol Pharm 2023; 20:1975-1989. [PMID: 36825806 DOI: 10.1021/acs.molpharmaceut.2c00897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Next-generation cancer immunotherapies may utilize immunostimulants to selectively activate the host immune system against tumor cells. Checkpoint inhibitors (CPIs) like anti-PD1/PDL-1 that inhibit immunosuppression have shown unprecedented success but are only effective in the 20-30% of patients that possess an already "hot" (immunogenic) tumor. In this regard, intratumoral (IT) injection of immunostimulants is a promising approach since they can work synergistically with CPIs to overcome the resistance to immunotherapies by inducing immune stimulation in the tumor. One such immunostimulant is granulocyte macrophage-colony-stimulating factor (GMCSF) that functions by recruiting and activating antigen-presenting cells (dendritic cells) in the tumor, thereby initiating anti-tumor immune responses. However, key problems with GMCSF are lack of efficacy and the risk of systemic toxicity caused by the leakage of GMCSF from the tumor tissue. We have designed tumor-retentive versions of GMCSF that are safe yet potent immunostimulants for the local treatment of solid tumors. The engineered GMCSFs (eGMCSF) were synthesized by recombinantly fusing tumor-ECM (extracellular matrix) binding peptides to GMCSF. The eGMCSFs exhibited enhanced tumor binding and potent immunological activity in vitro and in vivo. Upon IT administration, the tumor-retentive eGMCSFs persisted in the tumor, thereby alleviating systemic toxicity, and elicited localized immune activation to effectively turn an unresponsive immunologically "cold" tumor "hot".
Collapse
Affiliation(s)
| | - Chad E Groer
- HylaPharm, LLC, Lawrence, Kansas 66047, United States.,Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Huan Gong
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Vivian Yudistyra
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| | - Marcus Laird Forrest
- HylaPharm, LLC, Lawrence, Kansas 66047, United States.,Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Cory J Berkland
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66045, United States.,Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States.,Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
9
|
Burkard M, Niessner H, Leischner C, Piotrowsky A, Renner O, Marongiu L, Lauer UM, Busch C, Sinnberg T, Venturelli S. High-Dose Ascorbate in Combination with Anti-PD1 Checkpoint Inhibition as Treatment Option for Malignant Melanoma. Cells 2023; 12:254. [PMID: 36672190 PMCID: PMC9857291 DOI: 10.3390/cells12020254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Ascorbate acts as a prooxidant when administered parenterally at high supraphysiological doses, which results in the generation of hydrogen peroxide in dependence on oxygen. Most cancer cells are susceptible to the emerging reactive oxygen species (ROS). Accordingly, we evaluated high-dose ascorbate for the treatment of the B16F10 melanoma model. To investigate the effects of ascorbate on the B16F10 cell line in vitro, viability, cellular impedance, and ROS production were analyzed. In vivo, C57BL/6NCrl mice were subcutaneously injected into the right flank with B16F10 cells and tumor-bearing mice were treated intraperitoneally with ascorbate (3 g/kg bodyweight), immunotherapy (anti-programmed cell death protein 1 (PD1) antibody J43; 2 mg/kg bodyweight), or both treatments combined. The efficacy and toxicity were analyzed by measuring the respective tumor sizes and mouse weights accompanied by histological analysis of the protein levels of proliferating cell nuclear antigen (Pcna), glucose transporter 1 (Glut-1), and CD3. Treatment of B16F10 melanoma-carrying mice with high-dose ascorbate yielded plasma levels in the pharmacologically effective range, and ascorbate showed efficacy as a monotherapy and when combined with PD1 inhibition. Our data suggest the applicability of ascorbate as an additional therapeutic agent that can be safely combined with immunotherapy and has the potential to potentiate anti-PD1-based immune checkpoint blockades.
Collapse
Affiliation(s)
- Markus Burkard
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Heike Niessner
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
| | - Christian Leischner
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Alban Piotrowsky
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Olga Renner
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
| | - Luigi Marongiu
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Department of Internal Medicine VIII, University Hospital Tuebingen, Otfried-Mueller-Straße 10, 72076 Tuebingen, Germany
| | - Ulrich M. Lauer
- Department of Internal Medicine VIII, University Hospital Tuebingen, Otfried-Mueller-Straße 10, 72076 Tuebingen, Germany
| | - Christian Busch
- Dermatologie zum Delfin, Stadthausstraße 12, 8400 Winterthur, Switzerland
| | - Tobias Sinnberg
- Division of Dermatooncology, Department of Dermatology, University of Tuebingen, Liebermeisterstraße 25, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, 72076 Tuebingen, Germany
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Sascha Venturelli
- Department of Nutritional Biochemistry, Institute of Nutritional Sciences, University of Hohenheim, Garbenstraße 30, 70599 Stuttgart, Germany
- Department of Vegetative and Clinical Physiology, Institute of Physiology, University of Tuebingen, Wilhelmstraße 56, 72074 Tuebingen, Germany
| |
Collapse
|
10
|
Basudan AM. The Role of Immune Checkpoint Inhibitors in Cancer Therapy. Clin Pract 2022; 13:22-40. [PMID: 36648843 PMCID: PMC9844484 DOI: 10.3390/clinpract13010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Over the years, immune checkpoint inhibitors (CPIs) have become a powerful treatment strategy in the field of cancer immunotherapy. In the last decade, the number of FDA-approved CPIs has been increasing prominently, opening new horizons for the treatment of a wide range of tumor types. Pointedly, three immune checkpoint molecules have been under extensive research, which include cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein-1 (PD-1) and its ligand-1 (PD-L1). Despite remarkable success, not all patients respond positively to therapy, which highlights the complexity of the tumor microenvironment (TME) and immune system. This has led to the identification of molecular biomarkers to predict response and toxicity. In addition, there has been an emerging focus on developing new delivery and targeting approaches for better drug efficacy and potency. In this review, we highlight the mechanism of action of major CPIs, their clinical impact, variation in effectiveness, response prediction, updated clinical indications, current challenges and limitations, promising novel approaches, and future directions.
Collapse
Affiliation(s)
- Ahmed M Basudan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| |
Collapse
|
11
|
Huang S, Zhu Y, Zhang L, Zhang Z. Recent Advances in Delivery Systems for Genetic and Other Novel Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107946. [PMID: 34914144 DOI: 10.1002/adma.202107946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/11/2021] [Indexed: 06/14/2023]
Abstract
Vaccination is one of the most successful and cost-effective prophylactic measures against diseases, especially infectious diseases including smallpox and polio. However, the development of effective prophylactic or therapeutic vaccines for other diseases such as cancer remains challenging. This is often due to the imprecise control of vaccine activity in vivo which leads to insufficient/inappropriate immune responses or short immune memory. The development of new vaccine types in recent decades has created the potential for improving the protective potency against these diseases. Genetic and subunit vaccines are two major categories of these emerging vaccines. Owing to their nature, they rely heavily on delivery systems with various functions, such as effective cargo protection, immunogenicity enhancement, targeted delivery, sustained release of antigens, selective activation of humoral and/or cellular immune responses against specific antigens, and reduced adverse effects. Therefore, vaccine delivery systems may significantly affect the final outcome of genetic and other novel vaccines and are vital for their development. This review introduces these studies based on their research emphasis on functional design or administration route optimization, presents recent progress, and discusses features of new vaccine delivery systems, providing an overview of this field.
Collapse
Affiliation(s)
- Shiqi Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Yining Zhu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
12
|
Duhen T, Gough MJ, Leidner RS, Stanton SE. Development and therapeutic manipulation of the head and neck cancer tumor environment to improve clinical outcomes. FRONTIERS IN ORAL HEALTH 2022; 3:902160. [PMID: 35937775 PMCID: PMC9354490 DOI: 10.3389/froh.2022.902160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical response to cancer therapies involves the complex interplay between the systemic, tumoral, and stromal immune response as well as the direct impact of treatments on cancer cells. Each individual's immunological and cancer histories are different, and their carcinogen exposures may differ. This means that even though two patients with oral tumors may carry an identical mutation in TP53, they are likely to have different pre-existing immune responses to their tumors. These differences may arise due to their distinct accessory mutations, genetic backgrounds, and may relate to clinical factors including previous chemotherapy exposure and concurrent medical comorbidities. In isolation, their cancer cells may respond similarly to cancer therapy, but due to their baseline variability in pre-existing immune responses, patients can have different responses to identical therapies. In this review we discuss how the immune environment of tumors develops, the critical immune cell populations in advanced cancers, and how immune interventions can manipulate the immune environment of patients with pre-malignancies or advanced cancers to improve therapeutic outcomes.
Collapse
Affiliation(s)
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | | | | |
Collapse
|
13
|
Kumar A, Taghi Khani A, Sanchez Ortiz A, Swaminathan S. GM-CSF: A Double-Edged Sword in Cancer Immunotherapy. Front Immunol 2022; 13:901277. [PMID: 35865534 PMCID: PMC9294178 DOI: 10.3389/fimmu.2022.901277] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine that drives the generation of myeloid cell subsets including neutrophils, monocytes, macrophages, and dendritic cells in response to stress, infections, and cancers. By modulating the functions of innate immune cells that serve as a bridge to activate adaptive immune responses, GM-CSF globally impacts host immune surveillance under pathologic conditions. As with other soluble mediators of immunity, too much or too little GM-CSF has been found to promote cancer aggressiveness. While too little GM-CSF prevents the appropriate production of innate immune cells and subsequent activation of adaptive anti-cancer immune responses, too much of GM-CSF can exhaust immune cells and promote cancer growth. The consequences of GM-CSF signaling in cancer progression are a function of the levels of GM-CSF, the cancer type, and the tumor microenvironment. In this review, we first discuss the secretion of GM-CSF, signaling downstream of the GM-CSF receptor, and GM-CSF’s role in modulating myeloid cell homeostasis. We then outline GM-CSF’s anti-tumorigenic and pro-tumorigenic effects both on the malignant cells and on the non-malignant immune and other cells in the tumor microenvironment. We provide examples of current clinical and preclinical strategies that harness GM-CSF’s anti-cancer potential while minimizing its deleterious effects. We describe the challenges in achieving the Goldilocks effect during administration of GM-CSF-based therapies to patients with cancer. Finally, we provide insights into how technologies that map the immune microenvironment spatially and temporally may be leveraged to intelligently harness GM-CSF for treatment of malignancies.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Adeleh Taghi Khani
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Ashly Sanchez Ortiz
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Srividya Swaminathan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
- Department of Hematological Malignancies, Beckman Research Institute of City of Hope, Monrovia, CA, United States
- *Correspondence: Srividya Swaminathan,
| |
Collapse
|
14
|
Michielin O, Lalani AK, Robert C, Sharma P, Peters S. Defining unique clinical hallmarks for immune checkpoint inhibitor-based therapies. J Immunother Cancer 2022; 10:e003024. [PMID: 35078922 PMCID: PMC8796265 DOI: 10.1136/jitc-2021-003024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
IntroductionImmuno-oncology therapies, including immune checkpoint inhibitors (ICIs), have transformed cancer care and have brought into question whether classic oncology efficacy assessments adequately describe the distinctive responses observed with these agents. With more ICI-based therapies being approved across multiple tumor types, it is essential to define unique clinical hallmarks of these agents and their associated assessments to better reflect the therapeutic impact for both patients and physicians. Long-term survival and objective responses, such as depth and durability of responses, treatment-free survival, efficacy in brain metastases, improved health-related quality of life, and unique safety profiles, are among the hallmarks that have emerged for ICI therapies. An established clinical hallmark is a sustained long-term survival, as evidenced by a delayed separation of Kaplan-Meier survival curves, and a plateau at ~3 years. Combination ICI therapies provide the opportunity to raise this plateau, thereby affording durable survival benefits to more patients. Deepening of responses over time is a unique clinical ICI hallmark, with patients responding long term and with more durable complete responses. Depth of response has demonstrated prognostic value for long-term survival in some cancers, and several ICI studies have shown sustained responses even after discontinuing ICI therapy, offering the potential for treatment-free intervals. Although clinical evidence supporting efficacy in brain metastases is limited, favorable ICI intracranial responses have been seen that are largely concordant with extracranial responses. While patient outcomes can be significantly improved with ICIs, they are associated with unique immune-mediated adverse reactions (IMARs), including delayed ICI toxicities, and may require multidisciplinary management for optimal care. Interestingly, patients discontinuing ICIs for IMARs may maintain responses similar to patients who did not discontinue for an IMAR, whether they restarted ICI therapy or not.ConclusionHerein, we comprehensively review and refine the clinical hallmarks uniquely associated with ICI therapies, which not only will rejuvenate our assessment of ICI therapeutic outcomes but also will lead to a greater appreciation of the effectiveness of ICI therapies.
Collapse
Affiliation(s)
- Olivier Michielin
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Aly-Khan Lalani
- Department of Oncology, Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| | - Caroline Robert
- Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Padmanee Sharma
- Departments of Genitourinary Medical Oncology and Immunology, UT MD Anderson Cancer Center, Houston, Texas, USA
| | - Solange Peters
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
15
|
Makaremi S, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Sgambato A, Ghorbaninezhad F, Safarpour H, Argentiero A, Brunetti O, Bernardini R, Silvestris N, Baradaran B. Immune Checkpoint Inhibitors in Colorectal Cancer: Challenges and Future Prospects. Biomedicines 2021; 9:1075. [PMID: 34572263 PMCID: PMC8467932 DOI: 10.3390/biomedicines9091075] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is a new pillar of cancer therapy that provides novel opportunities to treat solid tumors. In this context, the development of new drugs targeting immune checkpoints is considered a promising approach in colorectal cancer (CRC) treatment because it can be induce specific and durable anti-cancer effects. Despite many advances in the immunotherapy of CRC, there are still limitations and obstacles to successful treatment. The immunosuppressive function of the tumor microenvironment (TME) is one of the causes of poor response to treatment in CRC patients. For this reason, checkpoint-blocking antibodies have shown promising outcomes in CRC patients by blocking inhibitory immune checkpoints and enhancing immune responses against tumors. This review summarizes recent advances in immune checkpoint inhibitors (ICIs), such as CTLA-4, PD-1, PD-L1, LAG-3, and TIM-3 in CRC, and it discusses various therapeutic strategies with ICIs, including the double blockade of ICIs, combination therapy of ICIs with other immunotherapies, and conventional treatments. This review also delineates a new hopeful path in the combination of anti-PD-1/anti-PD-L1 with other ICIs such as anti-CTLA-4, anti-LAG-3, and anti-TIM-3 for CRC treatment.
Collapse
Affiliation(s)
- Shima Makaremi
- Department of Immunology & Microbiology, School of Medicine, Arak University of Medical Sciences, Arak 3848176941, Iran;
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Alessandro Sgambato
- Istituto di Ricovero e Cura a Carattere Scientifico Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 5972362 Rome, Italy;
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 5972362 Rome, Italy
| | - Farid Ghorbaninezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Antonella Argentiero
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
| | - Oronzo Brunetti
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95121 Catania, Italy;
| | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
- Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| |
Collapse
|
16
|
Mihalik NE, Wen S, Driesschaert B, Eubank TD. Formulation and In Vitro Characterization of PLGA/PLGA-PEG Nanoparticles Loaded with Murine Granulocyte-Macrophage Colony-Stimulating Factor. AAPS PharmSciTech 2021; 22:191. [PMID: 34169366 DOI: 10.1208/s12249-021-02049-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) has demonstrated notable clinical activity in cancer immunotherapy, but it is limited by systemic toxicities, poor bioavailability, rapid clearance, and instability in vivo. Nanoparticles (NPs) may overcome these limitations and provide a mechanism for passive targeting of tumors. This study aimed to develop GM-CSF-loaded PLGA/PLGA-PEG NPs and evaluate them in vitro as a potential candidate for in vivo administration. NPs were created by a phase-separation technique that did not require toxic/protein-denaturing solvents or harsh agitation techniques and encapsulated GM-CSF in a more stable precipitated form. NP sizes were within 200 nm for enhanced permeability and retention (EPR) effect with negative zeta potentials, spherical morphology, and high entrapment efficiencies. The optimal formulation was identified by sustained release of approximately 70% of loaded GM-CSF over 24 h, alongside an average size of 143 ± 35 nm and entrapment efficiency of 84 ± 5%. These NPs were successfully freeze-dried in 5% (w/v) hydroxypropyl-β-cyclodextrin for long-term storage and further characterized. Bioactivity of released GM-CSF was determined by observing GM-CSF receptor activation on murine monocytes and remained fully intact. NPs were not cytotoxic to murine bone marrow-derived macrophages (BMDMs) at concentrations up to 1 mg/mL as determined by MTT and trypan blue exclusion assays. Lastly, NP components generated no significant transcription of inflammation-regulating genes from BMDMs compared to IFNγ+LPS "M1" controls. This report lays the preliminary groundwork to validate in vivo studies with GM-CSF-loaded PLGA/PEG-PLGA NPs for tumor immunomodulation. Overall, these data suggest that in vivo delivery will be well tolerated.
Collapse
|
17
|
Tarhini AA, Joshi I, Garner F. Sargramostim and immune checkpoint inhibitors: combinatorial therapeutic studies in metastatic melanoma. Immunotherapy 2021; 13:1011-1029. [PMID: 34157863 DOI: 10.2217/imt-2021-0119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The use of immune checkpoint inhibitors in patients with metastatic melanoma generates clinical benefit, including improved survival. Yet disease resistance and immune-related adverse events persist as unmet needs. Sargramostim, a yeast-derived recombinant human GM-CSF, has shown clinical activity against diverse solid tumors, including metastatic melanoma. Here we review the use of sargramostim for treatment of advanced melanoma. Potential sargramostim applications in melanoma draw on the unique ability of GM-CSF to link innate and adaptive immune responses. We review preclinical and translational data describing the mechanism of action of sargramostim and synergy with immune checkpoint inhibitors to enhance efficacy and reduce treatment-related toxicity.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Cutaneous Oncology & Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Ila Joshi
- Pre-Clinical & Translational Research & Development, Partner Therapeutics, 19 Muzzey Street, Lexington, MA 02421, USA
| | - Fiona Garner
- Immuno-Oncology Clinical Development & Translational Medicine, Partner Therapeutics, 19 Muzzey Street, Lexington, MA 02421, USA
| |
Collapse
|
18
|
Zhang N, Foiret J, Kheirolomoom A, Liu P, Feng Y, Tumbale S, Raie M, Wu B, Wang J, Fite BZ, Dai Z, Ferrara KW. Optimization of microbubble-based DNA vaccination with low-frequency ultrasound for enhanced cancer immunotherapy. ADVANCED THERAPEUTICS 2021; 4. [PMID: 34632048 DOI: 10.1002/adtp.202100033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immunotherapy is an important cancer treatment strategy; nevertheless, the lack of robust immune cell infiltration in the tumor microenvironment remains a factor in limiting patient response rates. In vivo gene delivery protocols can amplify immune responses and sensitize tumors to immunotherapies, yet non-viral transfection methods often sacrifice transduction efficiency for improved safety tolerance. To improve transduction efficiency, we optimized a strategy employing low ultrasound transmission frequency-induced bubble oscillation to introduce plasmids into tumor cells. Differential centrifugation isolated size-specific microbubbles. The diameter of the small microbubble population was 1.27 ± 0.89 μm and that of larger population was 4.23 ± 2.27 μm. Upon in vitro insonation with the larger microbubble population, 29.7% of cancer cells were transfected with DNA plasmids, higher than that with smaller microbubbles (18.9%, P <0.05) or positive control treatments with a commercial transfection reagent (12%, P < 0.01). After 48 h, gene expression increased more than two-fold in tumors treated with large, as compared with small, microbubbles. Furthermore, the immune response, including tumor infiltration of CD8+ T cells and F4/80+ macrophages, was enhanced. We believe that this safe and efficacious method can improve preclinical procedures and outcomes for DNA vaccines in cancer immunotherapy in the future.
Collapse
Affiliation(s)
- Nisi Zhang
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | | | - Pei Liu
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Yi Feng
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Spencer Tumbale
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Marina Raie
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Bo Wu
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - James Wang
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Zhifei Dai
- Department of Engineering, Peking University, Beijing, China
| | | |
Collapse
|
19
|
Campbell JR, McDonald BR, Mesko PB, Siemers NO, Singh PB, Selby M, Sproul TW, Korman AJ, Vlach LM, Houser J, Sambanthamoorthy S, Lu K, Hatcher SV, Lohre J, Jain R, Lan RY. Fc-Optimized Anti-CCR8 Antibody Depletes Regulatory T Cells in Human Tumor Models. Cancer Res 2021; 81:2983-2994. [PMID: 33757978 DOI: 10.1158/0008-5472.can-20-3585] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/07/2021] [Accepted: 03/15/2021] [Indexed: 11/16/2022]
Abstract
FOXP3+ regulatory T cells (Treg) play a critical role in mediating tolerance to self-antigens and can repress antitumor immunity through multiple mechanisms. Therefore, targeted depletion of tumor-resident Tregs is warranted to promote effective antitumor immunity while preserving peripheral homeostasis. Here, we propose the chemokine receptor CCR8 as one such optimal tumor Treg target. CCR8 was expressed by Tregs in both murine and human tumors, and unlike CCR4, a Treg depletion target in the clinic, CCR8 was selectively expressed on suppressive tumor Tregs and minimally expressed on proinflammatory effector T cells (Teff). Preclinical mouse tumor modeling showed that depletion of CCR8+ Tregs through an FcyR-engaging anti-CCR8 antibody, but not blockade, enabled dose-dependent, effective, and long-lasting antitumor immunity that synergized with PD-1 blockade. This depletion was tumor Treg-restricted, sparing CCR8+ T cells in the spleen, thymus, and skin of mice. Importantly, Fc-optimized, nonfucosylated (nf) anti-human CCR8 antibodies specifically depleted Tregs and not Teffs in ex vivo tumor cultures from primary human specimens. These findings suggest that anti-CCR8-nf antibodies may deliver optimal tumor-targeted Treg depletion in the clinic, providing long-term antitumor memory responses while limiting peripheral toxicities. SIGNIFICANCE: These findings show that selective depletion of regulatory T cells with an anti-CCR8 antibody can improve antitumor immune responses as a monotherapy or in combination with other immunotherapies. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/11/2983/F1.large.jpg.
Collapse
Affiliation(s)
| | | | | | | | | | - Mark Selby
- Bristol Myers Squibb, Redwood City, California
| | | | | | | | - Jeff Houser
- Bristol Myers Squibb, Redwood City, California
| | | | - Kai Lu
- Bristol Myers Squibb, Redwood City, California
| | | | - Jack Lohre
- Bristol Myers Squibb, Redwood City, California
| | - Renu Jain
- Bristol Myers Squibb, Redwood City, California.
| | - Ruth Y Lan
- Bristol Myers Squibb, Redwood City, California.
| |
Collapse
|
20
|
McGray AJR, Bernard D, Hallett R, Kelly R, Jha M, Gregory C, Bassett JD, Hassell JA, Pare G, Wan Y, Bramson JL. Combined vaccination and immunostimulatory antibodies provides durable cure of murine melanoma and induces transcriptional changes associated with positive outcome in human melanoma patients. Oncoimmunology 2021; 1:419-431. [PMID: 22754760 PMCID: PMC3382903 DOI: 10.4161/onci.19534] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We have developed a recombinant adenovirus vaccine encoding dopachrome tautomerase (rHuAd5-hDCT) that produces robust DCT-specific immunity, but only provides modest suppression of murine melanoma. In the current study, an agonist antibody against 4-1BB was shown to enhance rHuAd5-hDCT efficacy and evoke tumor regression, but most tumors ultimately relapsed. The vaccine triggered upregulation of the immune inhibitory PD-1 signaling pathway and PD-1 blockade dramatically enhanced the rHuAd5-hDCT + anti-4-1BB strategy, resulting in complete regression of growing tumors in > 70% of recipients. The impact of the combined anti-4-1BB/anti-PD-1 treatment did not manifest as a dramatic enhancement in either the magnitude or functionality of DCT-specific tumor infiltrating lymphocytes relative to either treatment alone. Rather, a synergistic enhancement in intratumoral cytokine expression was observed, suggesting that the benefit of the combined therapy was a local event within the tumor. Global transcriptional analysis revealed immunological changes within the tumor following the curative vaccination, which extended beyond the T cell compartment. We identified an immune signature of 85 genes associated with clearance of murine melanoma that correlated with improved survival outcome in two independent cohorts of human melanoma patients. Our data reinforce the concept that successful vaccination must overcome local hurdles in the tumor microenvironment that are not manifest within the periphery. Further, tumor rejection following vaccination involves more than simply T cells. Finally, the association of our immune signature with positive survival outcome in human melanoma patients suggests that similar vaccination strategies may be promising for melanoma treatment.
Collapse
Affiliation(s)
- A J Robert McGray
- Department of Pathology and Molecular Medicine; McMaster University; Hamilton, ON Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Verkleij CPM, Jhatakia A, Broekmans MEC, Frerichs KA, Zweegman S, Mutis T, Bezman NA, van de Donk NWCJ. Preclinical Rationale for Targeting the PD-1/PD-L1 Axis in Combination with a CD38 Antibody in Multiple Myeloma and Other CD38-Positive Malignancies. Cancers (Basel) 2020; 12:cancers12123713. [PMID: 33321969 PMCID: PMC7764511 DOI: 10.3390/cancers12123713] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022] Open
Abstract
Simple Summary The CD38-targeting antibody daratumumab mediates its anti-myeloma activities not only through direct effects on tumor cells, but also by its effects on T-cell immunity through depletion of CD38+ immune suppressor cells. We hypothesized that combining daratumumab with modulators of other potent immune inhibitory pathways, such as the PD-1/PD-L1 axis, may further improve its efficacy. We show that during MM progression there is increased expression of the PD-1/PD-L1 pathway components in the bone marrow microenvironment. Although nivolumab (a PD-1 checkpoint inhibitor) moderately increased T-cell frequencies in ex vivo experiments with bone marrow samples from MM patients, no single agent activity was observed, and addition of nivolumab did not enhance the activity of daratumumab in these short-term assays. However, with a longer treatment duration, in mouse experiments, we demonstrate that anti-CD38 and anti-PD-1 antibodies synergize to eradicate MM cells. In addition, our results suggest that this combined immunotherapeutic approach may also be beneficial in other CD38-positive malignancies. Abstract The CD38-targeting antibody daratumumab mediates its anti-myeloma activities not only through Fc-receptor-dependent effector mechanisms, but also by its effects on T-cell immunity through depletion of CD38+ regulatory T-cells, regulatory B-cells, and myeloid-derived suppressor cells. Therefore, combining daratumumab with modulators of other potent immune inhibitory pathways, such as the PD-1/PD-L1 axis, may further improve its efficacy. We show that multiple myeloma (MM) cells from relapsed/refractory patients have increased expression of PD-L1, compared to newly diagnosed patients. Furthermore, PD-1 is upregulated on T-cells from both newly diagnosed and relapsed/refractory MM patients, compared to healthy controls. In short-term experiments with bone marrow samples from MM patients, daratumumab-mediated lysis was mainly associated with the MM cells’ CD38 expression levels and the effector (NK-cells/monocytes/T-cells)-to-target ratio, but not with the PD-L1 expression levels or PD-1+ T-cell frequencies. Although PD-1 blockade with nivolumab did not affect MM cell viability or enhanced daratumumab-mediated lysis in short-term ex vivo experiments, nivolumab resulted in a mild but clear increase in T-cell numbers. Moreover, with a longer treatment duration, PD-1 blockade markedly improved anti-CD38 antibody-mediated cytotoxicity in vivo in murine CD38+ tumor models. In conclusion, dual targeting of CD38 and PD-1 may represent a promising strategy for treating MM and other CD38-positive malignancies.
Collapse
Affiliation(s)
- Christie P. M. Verkleij
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (C.P.M.V.); (M.E.C.B.); (K.A.F.); (S.Z.); (T.M.)
| | - Amy Jhatakia
- Bristol-Myers Squibb, Redwood City, CA 94063, USA; (A.J.); (N.A.B.)
| | - Marloes E. C. Broekmans
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (C.P.M.V.); (M.E.C.B.); (K.A.F.); (S.Z.); (T.M.)
| | - Kristine A. Frerichs
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (C.P.M.V.); (M.E.C.B.); (K.A.F.); (S.Z.); (T.M.)
| | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (C.P.M.V.); (M.E.C.B.); (K.A.F.); (S.Z.); (T.M.)
| | - Tuna Mutis
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (C.P.M.V.); (M.E.C.B.); (K.A.F.); (S.Z.); (T.M.)
| | - Natalie A. Bezman
- Bristol-Myers Squibb, Redwood City, CA 94063, USA; (A.J.); (N.A.B.)
- Arsenal Bio, San Francisco, CA 94080, USA
| | - Niels W. C. J. van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (C.P.M.V.); (M.E.C.B.); (K.A.F.); (S.Z.); (T.M.)
- Correspondence: ; Tel.: +31-(0)20-4442604
| |
Collapse
|
22
|
Gu Z, Da Silva CG, Van der Maaden K, Ossendorp F, Cruz LJ. Liposome-Based Drug Delivery Systems in Cancer Immunotherapy. Pharmaceutics 2020; 12:E1054. [PMID: 33158166 PMCID: PMC7694212 DOI: 10.3390/pharmaceutics12111054] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy has shown remarkable progress in recent years. Nanocarriers, such as liposomes, have favorable advantages with the potential to further improve cancer immunotherapy and even stronger immune responses by improving cell type-specific delivery and enhancing drug efficacy. Liposomes can offer solutions to common problems faced by several cancer immunotherapies, including the following: (1) Vaccination: Liposomes can improve the delivery of antigens and other stimulatory molecules to antigen-presenting cells or T cells; (2) Tumor normalization: Liposomes can deliver drugs selectively to the tumor microenvironment to overcome the immune-suppressive state; (3) Rewiring of tumor signaling: Liposomes can be used for the delivery of specific drugs to specific cell types to correct or modulate pathways to facilitate better anti-tumor immune responses; (4) Combinational therapy: Liposomes are ideal vehicles for the simultaneous delivery of drugs to be combined with other therapies, including chemotherapy, radiotherapy, and phototherapy. In this review, different liposomal systems specifically developed for immunomodulation in cancer are summarized and discussed.
Collapse
Affiliation(s)
- Zili Gu
- Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (Z.G.); (C.G.D.S.)
| | - Candido G. Da Silva
- Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (Z.G.); (C.G.D.S.)
| | - Koen Van der Maaden
- Tumor Immunology Group, Department of Immunology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (K.v.d.M.); (F.O.)
- TECOdevelopment GmbH, 53359 Rheinbach, Germany
| | - Ferry Ossendorp
- Tumor Immunology Group, Department of Immunology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (K.v.d.M.); (F.O.)
| | - Luis J. Cruz
- Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (Z.G.); (C.G.D.S.)
| |
Collapse
|
23
|
Plitnick LM, Hutchins B, Dubey S, Li N, Amin RP, Born S, Mangadu R, Phillips JH, Sriram V, Herzyk DJ. A T-cell-dependent antibody response study using a murine surrogate anti-PD-1 monoclonal antibody as an alternative to a non-human primate model. J Immunotoxicol 2020; 17:175-185. [PMID: 33078980 DOI: 10.1080/1547691x.2020.1826020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The programmed cell death 1 (PD-1) pathway represents a major immune checkpoint which may be engaged by cells in a tumor microenvironment to overcome active T-cell immune surveillance. Pembrolizumab (Keytruda®) is a potent and highly selective humanized monoclonal antibody (mAb) of the IgG4/κ isotype designed to directly block the interaction between PD-1 and its ligands, PD-L1 and PD-L2. The current work was focused on developing a mouse T-Dependent Antibody Response (TDAR) model using a murinized rat anti-mouse PD-1 antibody (muDX400; a rodent surrogate for pembrolizumab) to evaluate the potential impact of treatment with a PD-1 inhibitor on immune responses to an antigen challenge (e.g. HBsAg in Hepatitis B vaccine). Despite the lower binding affinity and T1/2 compared to pembrolizumab, ligand blocking data indicated muDX400 had appropriate pharmacological activity and demonstrated efficacy in mouse tumor models, thus was suitable for pharmacodynamic and vaccination studies in mice. In a vaccination study in which mice were concomitantly administered muDX400 and the Hepatitis B vaccine, muDX400 was well-tolerated and did not result in any immune-mediated adverse effects. The treatment with muDX400 was associated with a shift in the ratio between naive and memory cells in both CD4+ and CD8+ T-lymphocytes in the spleen but did not affect anti-HBsAg antibody response profile. The mouse TDAR model using the Hepatitis B vaccine and the surrogate anti-PD1 monoclonal antibody was a useful tool in the evaluation of the potential immune-mediated effects of pembrolizumab following vaccination and appears to be a suitable alternative for the nonhuman primate TDAR models utilized for other checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | | | - Nianyu Li
- Merck & Co., Inc, Kenilworth, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Kerr MD, McBride DA, Chumber AK, Shah NJ. Combining therapeutic vaccines with chemo- and immunotherapies in the treatment of cancer. Expert Opin Drug Discov 2020; 16:89-99. [PMID: 32867561 DOI: 10.1080/17460441.2020.1811673] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Breakthroughs in cancer immunotherapy have spurred interest in the development of vaccines to mediate prophylactic protection and therapeutic efficacy against primary tumors or to prevent relapse. However, immunosuppressive mechanisms employed by cancer cells to generate effective resistance have hampered clinical translation of therapeutic cancer vaccines. To enhance vaccine efficacy, the immunomodulatory properties of cytoreductive therapies could amplify a cancer-specific immune response. AREAS COVERED Herein, the authors discuss therapeutic cancer vaccines that harness whole cells and antigen-targeted vaccines. First, recent advancements in both autologous and allogeneic whole-cell vaccines and combinations with checkpoint blockade and chemotherapy are reviewed. Next, tumor antigen-targeted vaccines using peptide-based vaccines and DNA-vaccines are discussed. Finally, combination therapies using antigen-targeted vaccines are reviewed. EXPERT OPINION A deeper understanding of the immunostimulatory properties of cytoreductive therapies has supported their utility in combination therapies involving cancer vaccines as a potential strategy to induce a durable anti-tumor immune response for multiple types of cancers. Based on current evidence, combination therapies may have synergies that depend on the identity of the cytotoxic agent, vaccine target, dosing schedule, and cancer type. Together, these observations suggest that combining cancer vaccines with immunomodulatory cytoreductive therapy is a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Matthew D Kerr
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - David A McBride
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - Arun K Chumber
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - Nisarg J Shah
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA.,Program in Immunology, University of California , San Diego, CA, USA.,San Diego Center for Precision Immunotherapy, Moores Cancer Center, University of California , San Diego, CA, USA
| |
Collapse
|
25
|
Modulation of Determinant Factors to Improve Therapeutic Combinations with Immune Checkpoint Inhibitors. Cells 2020; 9:cells9071727. [PMID: 32707692 PMCID: PMC7408477 DOI: 10.3390/cells9071727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 01/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICPi) have shown their superiority over conventional therapies to treat some cancers. ICPi are effective against immunogenic tumors. However, patients with tumors poorly infiltrated with immune cells do not respond to ICPi. Combining ICPi with other anticancer therapies such as chemotherapy, radiation, or vaccines, which can stimulate the immune system and recruit antitumor T cells into the tumor bed, may be a relevant strategy to increase the proportion of responding patients. Such an approach still raises the following questions: What are the immunological features modulated by immunogenic therapies that can be critical to ensure not only immediate but also long-lasting tumor protection? How must the combined treatments be administered to the patients to harness their full potential while limiting adverse immunological events? Here, we address these points by reviewing how immunogenic anticancer therapies can provide novel therapeutic opportunities upon combination with ICPi. We discuss their ability to create a permissive tumor microenvironment through the generation of inflamed tumors and stimulation of memory T cells such as resident (TRM) and stem-cell like (TSCM) cells. We eventually underscore the importance of sequence, dose, and duration of the combined anticancer therapies to design optimal and successful cancer immunotherapy strategies.
Collapse
|
26
|
Prognostic and Clinicopathological Significance of PD-L1 in Patients with Cholangiocarcinoma: A Meta-Analysis. DISEASE MARKERS 2020; 2020:1817931. [PMID: 32724483 PMCID: PMC7381947 DOI: 10.1155/2020/1817931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 05/12/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022]
Abstract
Background In recent years, there is growing literature on the prognostic significance of programmed death-ligand 1 (PD-L1) in cholangiocarcinoma (CCA); however, data have been conflicting. Therefore, the objective of this study was to assess the correlation between PD-L1 and prognosis in CCA through meta-analysis. Methods Published studies were retrieved from the Web of Science, PubMed, Embase, and Cochrane Library up to April 17, 2020. The relationships between PD-L1 expression and survival outcomes were assessed using hazard ratios (HRs) and 95% confidence intervals (CIs). Results Eighteen studies consisting of 2012 patients were included. Overexpression of PD-L1 was significantly associated with worse overall survival (OS) (HR = 1.58, 95%CI = 1.30 - 1.92, p < 0.001) but not with poor disease-free survival (DFS) (HR = 1.03, 95%CI = 0.68 - 1.55, p = 0.895) in CCA. Moreover, PD-L1 was associated with low differentiation (OR = 1.43, 95%CI = 1.09 - 1.87, p = 0.010) and higher pN stage (OR = 1.45, 95%CI = 1.10 - 1.92, p = 0.009) but not with sex, TNM stage, vascular invasion, perineural invasion, age, or tumor size. Conclusion High PD-L1 expression was associated with worse OS, poor differentiation, and higher pN stage in patients with CCA. PD-L1 could be a potential prognostic marker in CCA.
Collapse
|
27
|
Immunotherapy in Prostate Cancer. Cancers (Basel) 2020; 12:cancers12071752. [PMID: 32630247 PMCID: PMC7409298 DOI: 10.3390/cancers12071752] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 01/03/2023] Open
Abstract
Immunotherapy encompasses a wide range of therapies to engage the immune system to target malignancies. In recent years, immunotherapy has made a major impact on treatment of metastatic cancer and has altered standard of care for many tumor types. However, predicting and understanding responses across tumor types has been challenging. While some metastatic cancers have shown dramatic responses to immunotherapy, such as melanoma, lung cancer, and renal cell carcinoma, prostate cancer has generally failed to show a significant response. However, small series of prostate cancer patients have shown impressive responses to cellular and immunotherapy. This review summarizes the current data for immunotherapy’s use in prostate cancer, as well as how currently available data might help predict patient responses to immunotherapy. Specifically, we will review vaccine-based therapies, immune checkpoint inhibitors, and future directions that are actively being explored.
Collapse
|
28
|
Roy S, Sethi TK, Taylor D, Kim YJ, Johnson DB. Breakthrough concepts in immune-oncology: Cancer vaccines at the bedside. J Leukoc Biol 2020; 108:1455-1489. [PMID: 32557857 DOI: 10.1002/jlb.5bt0420-585rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 12/11/2022] Open
Abstract
Clinical approval of the immune checkpoint blockade (ICB) agents for multiple cancer types has reinvigorated the long-standing work on cancer vaccines. In the pre-ICB era, clinical efforts focused on the Ag, the adjuvants, the formulation, and the mode of delivery. These translational efforts on therapeutic vaccines range from cell-based (e.g., dendritic cells vaccine Sipuleucel-T) to DNA/RNA-based platforms with various formulations (liposome), vectors (Listeria monocytogenes), or modes of delivery (intratumoral, gene gun, etc.). Despite promising preclinical results, cancer vaccine trials without ICB have historically shown little clinical activity. With the anticipation and expansion of combinatorial immunotherapeutic trials with ICB, the cancer vaccine field has entered the personalized medicine arena with recent advances in immunogenic neoantigen-based vaccines. In this article, we review the literature to organize the different cancer vaccines in the clinical space, and we will discuss their advantages, limits, and recent progress to overcome their challenges. Furthermore, we will also discuss recent preclinical advances and clinical strategies to combine vaccines with checkpoint blockade to improve therapeutic outcome and present a translational perspective on future directions.
Collapse
Affiliation(s)
- Sohini Roy
- Department of Otolaryngology - Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tarsheen K Sethi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David Taylor
- Department of Otolaryngology - Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Young J Kim
- Department of Otolaryngology - Head & Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
29
|
Trojaniello C, Vitale MG, Scarpato L, Esposito A, Ascierto PA. Melanoma immunotherapy: strategies to overcome pharmacological resistance. Expert Rev Anticancer Ther 2020; 20:289-304. [PMID: 32195606 DOI: 10.1080/14737140.2020.1745634] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Introduction: Although checkpoint inhibitors have provided a breakthrough in how melanoma is treated, about half of patients still do not respond due to primary or acquired resistance. New strategies are, therefore, required to increase the number of patients benefiting from immunotherapy. This systematic review investigates novel combinations that may overcome immune resistance in patients with melanoma.Areas covered: We provide an overview of immune-related resistance mechanisms and the various therapeutic strategies that can be considered in attempting to overcome these barriers, including combined immunotherapy approaches and combinations with chemotherapy, radiotherapy, and targeted therapy.Expert opinion: The immune response is a dynamic process in which the tumor microenvironment and immune cells interact in a variety of ways. New treatment approaches aim to enrich the tumor microenvironment with immune-infiltrate and increase response to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Claudia Trojaniello
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | | | - Luigi Scarpato
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Assunta Esposito
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
30
|
Jiao Q, Ren Y, Ariston Gabrie AN, Wang Q, Wang Y, Du L, Liu X, Wang C, Wang YS. Advances of immune checkpoints in colorectal cancer treatment. Biomed Pharmacother 2020; 123:109745. [DOI: 10.1016/j.biopha.2019.109745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/30/2022] Open
|
31
|
Kersch CN, Claunch CJ, Ambady P, Bucher E, Schwartz DL, Barajas RF, Iliff JJ, Risom T, Heiser L, Muldoon LL, Korkola JE, Gray JW, Neuwelt EA. Transcriptional signatures in histologic structures within glioblastoma tumors may predict personalized drug sensitivity and survival. Neurooncol Adv 2020; 2:vdaa093. [PMID: 32904984 PMCID: PMC7462280 DOI: 10.1093/noajnl/vdaa093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Glioblastoma is a rapidly fatal brain cancer that exhibits extensive intra- and intertumoral heterogeneity. Improving survival will require the development of personalized treatment strategies that can stratify tumors into subtypes that differ in therapeutic vulnerability and outcomes. Glioblastoma stratification has been hampered by intratumoral heterogeneity, limiting our ability to compare tumors in a consistent manner. Here, we develop methods that mitigate the impact of intratumoral heterogeneity on transcriptomic-based patient stratification. METHODS We accessed open-source transcriptional profiles of histological structures from 34 human glioblastomas from the Ivy Glioblastoma Atlas Project. Principal component and correlation network analyses were performed to assess sample inter-relationships. Gene set enrichment analysis was used to identify enriched biological processes and classify glioblastoma subtype. For survival models, Cox proportional hazards regression was utilized. Transcriptional profiles from 156 human glioblastomas were accessed from The Cancer Genome Atlas to externally validate the survival model. RESULTS We showed that intratumoral histologic architecture influences tumor classification when assessing established subtyping and prognostic gene signatures, and that indiscriminate sampling can produce misleading results. We identified the cellular tumor as a glioblastoma structure that can be targeted for transcriptional analysis to more accurately stratify patients by subtype and prognosis. Based on expression from cellular tumor, we created an improved risk stratification gene signature. CONCLUSIONS Our results highlight that biomarker performance for diagnostics, prognostics, and prediction of therapeutic response can be improved by analyzing transcriptional profiles in pure cellular tumor, which is a critical step toward developing personalized treatment for glioblastoma.
Collapse
Affiliation(s)
- Cymon N Kersch
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health and Science University, Portland, Oregon, USA
| | - Cheryl J Claunch
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Prakash Ambady
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health and Science University, Portland, Oregon, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Daniel L Schwartz
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, Oregon, USA
- Department of Neurology, Layton Aging and Alzheimer’s Disease Center, Oregon Health and Science University, Portland, Oregon, USA
| | - Ramon F Barajas
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, Oregon, USA
- Department of Radiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Jeffrey J Iliff
- Department of Neurology and Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Tyler Risom
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Laura Heiser
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Leslie L Muldoon
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health and Science University, Portland, Oregon, USA
| | - James E Korkola
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Joe W Gray
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Edward A Neuwelt
- Department of Neurology, Blood-Brain Barrier Program, Oregon Health and Science University, Portland, Oregon, USA
- Department of Neurosurgery, Oregon Health and Science University, Portland, Oregon, USA
- Office of Research and Development, Department of Veterans Affairs Medical Center, Portland, Oregon, USA
| |
Collapse
|
32
|
Survival Comparison between Melanoma Patients Treated with Patient-Specific Dendritic Cell Vaccines and Other Immunotherapies Based on Extent of Disease at the Time of Treatment. Biomedicines 2019; 7:biomedicines7040080. [PMID: 31614482 PMCID: PMC6966441 DOI: 10.3390/biomedicines7040080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 11/16/2022] Open
Abstract
Encouraging survival was observed in single arm and randomized phase 2 trials of patient-specific dendritic cell vaccines presenting autologous tumor antigens from autologous cancer cells that were derived from surgically resected metastases whose cells were self-renewing in vitro. Based on most advanced clinical stage and extent of tumor at the time of treatment, survival was best in patients classified as recurrent stage 3 without measurable disease. Next best was in stage 4 without measurable disease, and the worst survival was for measurable stage 4 disease. In this study, the survival of these patients was compared to the best contemporary controls that were gleaned from the clinical trial literature. The most comparable controls typically were from clinical trials testing other immunotherapy approaches. Even though contemporary controls typically had better prognostic features, median and/or long-term survival was consistently better in patients treated with this dendritic cell vaccine, except when compared to anti-programmed death molecule 1 (anti-PD-1). The clinical benefit of this patient-specific vaccine appears superior to a number of other immunotherapy approaches, but it is more complex to deliver than anti-PD-1 while equally effective. However, there is a strong rationale for combining such a product with anti-PD-1 in the treatment of patients with metastatic melanoma.
Collapse
|
33
|
Wang R, Gao C, Raymond M, Dito G, Kabbabe D, Shao X, Hilt E, Sun Y, Pak I, Gutierrez M, Melero I, Spreafico A, Carvajal RD, Ong M, Olszanski AJ, Milburn C, Thudium K, Yang Z, Feng Y, Fracasso PM, Korman AJ, Aanur P, Huang SMA, Quigley M. An Integrative Approach to Inform Optimal Administration of OX40 Agonist Antibodies in Patients with Advanced Solid Tumors. Clin Cancer Res 2019; 25:6709-6720. [PMID: 31573956 DOI: 10.1158/1078-0432.ccr-19-0526] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/29/2019] [Accepted: 08/09/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE The success of checkpoint blockade has led to a significant increase in the development of a broad range of immunomodulatory molecules for the treatment of cancer, including agonists against T-cell costimulatory receptors, such as OX40. Unlike checkpoint blockade, where complete and sustained receptor saturation may be required for maximal activity, the optimal dosing regimen and receptor occupancy for agonist agents is less well understood and requires further study. EXPERIMENTAL DESIGN We integrated both preclinical and clinical biomarker data sets centered on dose, exposure, receptor occupancy, receptor engagement, and downstream pharmacodynamic changes to model the optimal dose and schedule for the OX40 agonist antibody BMS-986178 alone and in combination with checkpoint blockade. RESULTS Administration of the ligand-blocking anti-mouse surrogate antibody OX40.23 or BMS-986178 as monotherapy or in combination with checkpoint blockade led to increased peripheral CD4+ and CD8+ T-cell activation in tumor-bearing mice and patients with solid tumors, respectively. OX40 receptor occupancy between 20% and 50% both in vitro and in vivo was associated with maximal enhancement of T-cell effector function by anti-OX40 treatment, whereas a receptor occupancy > 40% led to a profound loss in OX40 receptor expression, with clear implications for availability for repeat dosing. CONCLUSIONS Our results highlight the value of an integrated translational approach applied during early clinical development to aggregate preclinical and clinical data in an effort to define the optimal dose and schedule for T-cell agonists in the clinic.
Collapse
Affiliation(s)
- Rui Wang
- Sanofi, Oncology Clinical Translational Medicine, Cambridge, Massachusetts
| | - Chan Gao
- Bristol-Myers Squibb, Discovery Oncology, Redwood City, California
| | - Megan Raymond
- Bristol-Myers Squibb, Discovery Oncology, Redwood City, California
| | | | | | - Xiao Shao
- Bristol-Myers Squibb, Princeton, New Jersey
| | - Ed Hilt
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | - Irene Pak
- Bristol-Myers Squibb, Princeton, New Jersey
| | - Martin Gutierrez
- Divisions of Thoracic Oncology and Gastrointestinal Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, New Jersey
| | - Ignacio Melero
- Immunology and Immunotherapy Service, Center for Applied Medical Research, Clinica Universidad de Navarra, Pamplona, Spain
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada.,Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Richard D Carvajal
- Department of Medicine, Division of Hematology/Oncology, Columbia University Medical Center, New York, New York
| | - Michael Ong
- Department of Medicine, Division of Medical Oncology, University of Ottawa, Ottawa, Ontario, Canada
| | - Anthony J Olszanski
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple University, Philadelphia, Pennsylvania
| | | | | | - Zheng Yang
- Bristol-Myers Squibb, Princeton, New Jersey
| | - Yan Feng
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | - Alan J Korman
- Bristol-Myers Squibb, Discovery Oncology, Redwood City, California
| | | | | | - Michael Quigley
- Bristol-Myers Squibb, Discovery Oncology, Redwood City, California.
| |
Collapse
|
34
|
Tuo Z, Zong Y, Li J, Xiao G, Zhang F, Li G, Wang S, Lv Y, Xia J, Liu J. PD-L1 regulation by SDH5 via β-catenin/ZEB1 signaling. Oncoimmunology 2019; 8:1655361. [PMID: 31741753 PMCID: PMC6844322 DOI: 10.1080/2162402x.2019.1655361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 08/02/2019] [Accepted: 08/09/2019] [Indexed: 01/29/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a crucial target for lung cancer immunotherapy. In lung cancer patients with high PD-L1 expression, blocking or reducing its expression can inhibit tumor growth. PD-L1 is regulated by signaling pathways, transcription factors and epigenetic factors, such as the GSK3β/β-catenin pathway, P53 protein and EMT. In our previous study, succinate dehydrogenase 5 (SDH5) was reported to regulate ZEB1 expression, induce EMT and lead to lung cancer metastasis via the GSK3β/β-catenin pathway. It is possible that SDH5 is involved in the mechanisms of PD-L1 regulation.In the present study, we observed a negative correlation between the expression of PD-L1 and SDH5 in vivo and in vitro. The examination of patient tissues also confirmed our results. Furthermore, we also found that SDH5 could reverse PD-L1 expression by the GSK3β/β-catenin/ZEB1 pathways. All these results reveal that SDH5 regulates PD-L1 expression and suggest that SDH5 can be used as a marker to predict tumor immune micro-states and provide guidance for clinical immunotherapy.
Collapse
Affiliation(s)
- Zhan Tuo
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangqin Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Furong Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guiling Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sihua Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Lv
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Payandeh Z, Yarahmadi M, Nariman-Saleh-Fam Z, Tarhriz V, Islami M, Aghdam AM, Eyvazi S. Immune therapy of melanoma: Overview of therapeutic vaccines. J Cell Physiol 2019; 234:14612-14621. [PMID: 30706472 DOI: 10.1002/jcp.28181] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Melanoma is the most serious type of skin cancer which develops from the occurrence of genetic mutations in the melanocytes. Based on the features of melanoma tumors such as location, genetic profile and stage, there are several therapeutic strategies including surgery, chemotherapy, and radiotherapy. However, because of the appearance resistance mechanisms, the efficiency of these treatments strategies may be reduced. It has been demonstrated that therapeutic monoclonal antibodies can improve the efficiency of melanoma therapies. Recently, several mAbs, such as nivolumab, pembrolizumab, and ipilimumab, were approved for the immunotherapy of melanoma. The antibodies inhibit immune checkpoint receptors such as CTL4 and pd-1. Another therapeutic strategy for the treatment of melanoma is cancer vaccines, which improve clinical outcomes in patients. The combination therapy using antibodies and gene vaccine give us a new perspective in the treatment of melanoma patients. Herein, we present the recent progressions in the melanoma immunotherapy, especially dendritic cells mRNA vaccines by reviewing recent literature.
Collapse
Affiliation(s)
- Zahra Payandeh
- Immunology Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maral Yarahmadi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Islami
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Shirin Eyvazi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Biotechnology Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Newton JM, Hanoteau A, Liu HC, Gaspero A, Parikh F, Gartrell-Corrado RD, Hart TD, Laoui D, Van Ginderachter JA, Dharmaraj N, Spanos WC, Saenger Y, Young S, Sikora AG. Immune microenvironment modulation unmasks therapeutic benefit of radiotherapy and checkpoint inhibition. J Immunother Cancer 2019; 7:216. [PMID: 31409394 PMCID: PMC6693252 DOI: 10.1186/s40425-019-0698-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) for solid tumors, including those targeting programmed cell death 1 (PD-1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), have shown impressive clinical efficacy, however, most patients do not achieve durable responses. One major therapeutic obstacle is the immunosuppressive tumor immune microenvironment (TIME). Thus, we hypothesized that a strategy combining tumor-directed radiation with TIME immunomodulation could improve ICI response rates in established solid tumors. METHODS Using a syngeneic mouse model of human papillomavirus (HPV)-associated head and neck cancer, mEER, we developed a maximally effective regimen combining PD-1 and CTLA-4 inhibition, tumor-directed radiation, and two existing immunomodulatory drugs: cyclophosphamide (CTX) and a small-molecule inducible nitric oxide synthase (iNOS) inhibitor, L-n6-(1-iminoethyl)-lysine (L-NIL). We compared the effects of the various combinations of this regimen on tumor growth, overall survival, establishment of immunologic memory, and immunologic changes with flow cytometry and quantitative multiplex immunofluorescence. RESULTS We found PD-1 and CTLA-4 blockade, and radiotherapy alone or in combination, incapable of clearing established tumors or reversing the unfavorable balance of effector to suppressor cells in the TIME. However, modulation of the TIME with cyclophosphamide (CTX) and L-NIL in combination with dual checkpoint inhibition and radiation led to rejection of over 70% of established mEER tumors and doubled median survival in the B16 melanoma model. Anti-tumor activity was CD8+ T cell-dependent and led to development of immunologic memory against tumor-associated HPV antigens. Immune profiling revealed that CTX/L-NIL induced remodeling of myeloid cell populations in the TIME and tumor-draining lymph node and drove subsequent activation and intratumoral infiltration of CD8+ effector T cells. CONCLUSIONS Overall, this study demonstrates that modulation of the immunosuppressive TIME is required to unlock the benefits of ICIs and radiotherapy to induce immunologic rejection of treatment-refractory established solid tumors.
Collapse
Affiliation(s)
- Jared M. Newton
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Houston, TX USA
| | - Aurelie Hanoteau
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX USA
| | - Hsuan-Chen Liu
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Houston, TX USA
| | - Angelina Gaspero
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX USA
| | - Falguni Parikh
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX USA
| | - Robyn D. Gartrell-Corrado
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Columbia University Irving Medical Center/New York Presbyterian, New York, NY USA
| | - Thomas D. Hart
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center/New York Presbyterian, New York, NY USA
| | - Damya Laoui
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A. Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
| | - Neeraja Dharmaraj
- Department of Oral and Maxillofacial Surgery, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - William C. Spanos
- Department of Surgery, University of South Dakota, Sanford School of Medicine, Vermillion, SD USA
| | - Yvonne Saenger
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center/New York Presbyterian, New York, NY USA
| | - Simon Young
- Department of Oral and Maxillofacial Surgery, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Andrew G. Sikora
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX USA
- Department of Cell and Gene Therapy, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
37
|
Di Vito C, Mikulak J, Zaghi E, Pesce S, Marcenaro E, Mavilio D. NK cells to cure cancer. Semin Immunol 2019; 41:101272. [PMID: 31085114 DOI: 10.1016/j.smim.2019.03.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 12/12/2022]
Abstract
Natural Killer (NK) cells are innate lymphocytes able to mediate immune-surveillance and clearance of viral infected and tumor-transformed cells. Growing experimental and clinical evidence highlighted a dual role of NK cells either in the control of cancer development/progression or in promoting the onset of immune-suppressant tumor microenvironments. Indeed, several mechanisms of NK cell-mediated tumor escape have been described and these includes cancer-induced aberrant expression of activating and inhibitory receptors (i.e. NK cell immune checkpoints), impairments of NK cell migration to tumor sites and altered NK cell effector-functions. These phenomena highly contribute to tumor progression and metastasis formation. In this review, we discuss the latest insights on those NK cell receptors and related molecules that are currently being implemented in clinics either as possible prognostic factors or therapeutic targets to unleash NK cell anti-tumor effector-functions in vivo. Moreover, we address here the major recent advances in regard to the genetic modification and ex vivo expansion of anti-tumor specific NK cells used in innovative adoptive cellular transfer approaches.
Collapse
Affiliation(s)
- Clara Di Vito
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Joanna Mikulak
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Italy
| | - Elisa Zaghi
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy.
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Italy.
| |
Collapse
|
38
|
Kasikara C, Davra V, Calianese D, Geng K, Spires TE, Quigley M, Wichroski M, Sriram G, Suarez-Lopez L, Yaffe MB, Kotenko SV, De Lorenzo MS, Birge RB. Pan-TAM Tyrosine Kinase Inhibitor BMS-777607 Enhances Anti–PD-1 mAb Efficacy in a Murine Model of Triple-Negative Breast Cancer. Cancer Res 2019; 79:2669-2683. [DOI: 10.1158/0008-5472.can-18-2614] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/14/2019] [Accepted: 03/12/2019] [Indexed: 11/16/2022]
|
39
|
Margolis N, Markovits E, Markel G. Reprogramming lymphocytes for the treatment of melanoma: From biology to therapy. Adv Drug Deliv Rev 2019; 141:104-124. [PMID: 31276707 DOI: 10.1016/j.addr.2019.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/31/2019] [Accepted: 06/24/2019] [Indexed: 12/15/2022]
Abstract
This decade has introduced drastic changes in melanoma therapy, predominantly due to the materialization of the long promise of immunotherapy. Cytotoxic T cells are the chief component of the immune system, which are targeted by different strategies aimed to increase their capacity against melanoma cells. To this end, reprogramming of T cells occurs by T cell centered manipulation, targeting the immunosuppressive tumor microenvironment or altering the whole patient. These are enabled by delivery of small molecules, functional monoclonal antibodies, different subunit vaccines, as well as living lymphocytes, native or genetically engineered. Current FDA-approved therapies are focused on direct T cell manipulation, such as immune checkpoint inhibitors blocking CTLA-4 and/or PD-1, which paves the way for an effective immunotherapy backbone available for combination with other modalities. Here we review the biology and clinical developments that enable melanoma immunotherapy today and in the future.
Collapse
|
40
|
Kim IK, Koh CH, Jeon I, Shin KS, Kang TS, Bae EA, Seo H, Ko HJ, Kim BS, Chung Y, Kang CY. GM-CSF Promotes Antitumor Immunity by Inducing Th9 Cell Responses. Cancer Immunol Res 2019; 7:498-509. [PMID: 30728152 DOI: 10.1158/2326-6066.cir-18-0518] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/09/2018] [Accepted: 01/28/2019] [Indexed: 11/16/2022]
Abstract
GM-CSF as an adjuvant has been shown to promote antitumor immunity in mice and humans; however, the underlying mechanism of GM-CSF-induced antitumor immunity remains incompletely understood. In this study, we demonstrate that GM-CSF potentiates the efficacy of cancer vaccines through IL9-producing Th (Th9) cells. GM-CSF selectively enhanced Th9 cell differentiation by regulating the COX2-PGE2 pathway while inhibiting the differentiation of induced regulatory T (iTreg) cells in vitro and in vivo GM-CSF-activated monocyte-derived dendritic cells converted tumor-specific naïve Th cells into Th9 cells, and delayed tumor growth by inducing antitumor CTLs in an IL9-dependent manner. Our findings reveal a mechanism for the adjuvanticity of GM-CSF and provide a rationale for the use of GM-CSF in cancer vaccines.
Collapse
Affiliation(s)
- Il-Kyu Kim
- Laboratory of Immunology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Choong-Hyun Koh
- Laboratory of Immunology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Insu Jeon
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Kwang-Soo Shin
- Laboratory of Immunology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Tae-Seung Kang
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Eun-Ah Bae
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hyungseok Seo
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hyun-Ja Ko
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Republic of Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Byung-Seok Kim
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Chang-Yuil Kang
- Laboratory of Immunology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea. .,Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
41
|
Shen Z, Gu L, Mao D, Chen M, Jin R. Clinicopathological and prognostic significance of PD-L1 expression in colorectal cancer: a systematic review and meta-analysis. World J Surg Oncol 2019; 17:4. [PMID: 30609938 PMCID: PMC6320581 DOI: 10.1186/s12957-018-1544-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To analyze the prognostic value of programmed death factor ligand 1 (PD-L1) in colorectal cancer. METHODS Electronic databases, such as PubMed, Web of Science, Embase, and Cochrane library, were searched to identify studies evaluating the PD-L1 expression and overall survival (OS) in these patients. Afterwards, the relevant data were extracted to perform the meta-analysis. RESULTS A total of 3481 patients were included in 10 studies. The combined hazard ratio (HR) was 1.22 (95%CI = 1.01-1.48, P = 0.04), indicating that high expression of PD-L1 was significantly correlated with poor prognosis of colorectal cancer. Apropos of clinicopathological features, the merged odds ratio (OR) exhibited that highly expressed PD-L1 was firmly related to lymphatic invasion (OR = 3.49, 95%CI = 1.54-7.90, P = 0.003) and advanced stage (OR = 1.77, 95%CI = 1.41-2.23, P < 0.00001), but not correlative with patients' gender, microsatellite instability, or tumor location. CONCLUSION The expression of PD-L1 can be utilized as an independent factor in judging the prognosis of colorectal cancer, and patients with advanced cancer or lymphatic invasion are more likely to express PD-L1. This conclusion may lay a theoretical foundation for the application of PD-1/PD-L1 immunoassay point inhibitors but still needs verifying by sizeable well-designed cohort studies.
Collapse
Affiliation(s)
- Zefeng Shen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang China
| | - Lihu Gu
- Department of General Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang China
| | - Danyi Mao
- Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang China
| | - Manman Chen
- Affiliated Hospital of Medical School Ningbo University and Ningbo City Third Hospital, Ningbo, Zhejiang China
| | - Rongjia Jin
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang China
| |
Collapse
|
42
|
Kugel CH, Douglass SM, Webster MR, Kaur A, Liu Q, Yin X, Weiss SA, Darvishian F, Al-Rohil RN, Ndoye A, Behera R, Alicea GM, Ecker BL, Fane M, Allegrezza MJ, Svoronos N, Kumar V, Wang DY, Somasundaram R, Hu-Lieskovan S, Ozgun A, Herlyn M, Conejo-Garcia JR, Gabrilovich D, Stone EL, Nowicki TS, Sosman J, Rai R, Carlino MS, Long GV, Marais R, Ribas A, Eroglu Z, Davies MA, Schilling B, Schadendorf D, Xu W, Amaravadi RK, Menzies AM, McQuade JL, Johnson DB, Osman I, Weeraratna AT. Age Correlates with Response to Anti-PD1, Reflecting Age-Related Differences in Intratumoral Effector and Regulatory T-Cell Populations. Clin Cancer Res 2018; 24:5347-5356. [PMID: 29898988 PMCID: PMC6324578 DOI: 10.1158/1078-0432.ccr-18-1116] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/13/2018] [Accepted: 05/03/2018] [Indexed: 12/22/2022]
Abstract
Purpose: We have shown that the aged microenvironment increases melanoma metastasis, and decreases response to targeted therapy, and here we queried response to anti-PD1.Experimental Design: We analyzed the relationship between age, response to anti-PD1, and prior therapy in 538 patients. We used mouse models of melanoma, to analyze the intratumoral immune microenvironment in young versus aged mice and confirmed our findings in human melanoma biopsies.Results: Patients over the age of 60 responded more efficiently to anti-PD-1, and likelihood of response to anti-PD-1 increased with age, even when we controlled for prior MAPKi therapy. Placing genetically identical tumors in aged mice (52 weeks) significantly increased their response to anti-PD1 as compared with the same tumors in young mice (8 weeks). These data suggest that this increased response in aged patients occurs even in the absence of a more complex mutational landscape. Next, we found that young mice had a significantly higher population of regulatory T cells (Tregs), skewing the CD8+:Treg ratio. FOXP3 staining of human melanoma biopsies revealed similar increases in Tregs in young patients. Depletion of Tregs using anti-CD25 increased the response to anti-PD1 in young mice.Conclusions: While there are obvious limitations to our study, including our inability to conduct a meta-analysis due to a lack of available data, and our inability to control for mutational burden, there is a remarkable consistency in these data from over 500 patients across 8 different institutes worldwide. These results stress the importance of considering age as a factor for immunotherapy response. Clin Cancer Res; 24(21); 5347-56. ©2018 AACR See related commentary by Pawelec, p. 5193.
Collapse
Affiliation(s)
| | | | | | - Amanpreet Kaur
- The Wistar Institute, Philadelphia, Philadelphia
- University of the Sciences, Philadelphia, Philadelphia
| | - Qin Liu
- The Wistar Institute, Philadelphia, Philadelphia
| | - Xiangfan Yin
- The Wistar Institute, Philadelphia, Philadelphia
| | - Sarah A Weiss
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Farbod Darvishian
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Rami N Al-Rohil
- Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Abibatou Ndoye
- The Wistar Institute, Philadelphia, Philadelphia
- University of the Sciences, Philadelphia, Philadelphia
| | - Reeti Behera
- The Wistar Institute, Philadelphia, Philadelphia
| | - Gretchen M Alicea
- The Wistar Institute, Philadelphia, Philadelphia
- University of the Sciences, Philadelphia, Philadelphia
| | | | | | | | | | - Vinit Kumar
- The Wistar Institute, Philadelphia, Philadelphia
| | - Daniel Y Wang
- Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | | | - Siwen Hu-Lieskovan
- Department of Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | - Alpaslan Ozgun
- Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, Florida
| | | | | | | | | | - Theodore S Nowicki
- Department of Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | - Jeffrey Sosman
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Rajat Rai
- Melanoma Institute Australia and The University of Sydney, Westmead and Blacktown Hospitals Sydney, New South Wales, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia and The University of Sydney, Westmead and Blacktown Hospitals Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia and The University of Sydney, Westmead and Blacktown Hospitals Sydney, New South Wales, Australia
| | - Richard Marais
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Antoni Ribas
- Department of Medicine, University of California Los Angeles (UCLA), Los Angeles, California
| | - Zeynep Eroglu
- Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, Florida
| | - Michael A Davies
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Wurzburg, Wurzburg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, West German Cancer Center, University Duisburg-Essen, Essen, Germany
| | - Wei Xu
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ravi K Amaravadi
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander M Menzies
- Melanoma Institute Australia and The University of Sydney, Westmead and Blacktown Hospitals Sydney, New South Wales, Australia
| | | | - Douglas B Johnson
- Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Iman Osman
- Department of Medicine, New York University School of Medicine, New York, New York
| | | |
Collapse
|
43
|
Zhang X, Hu F, Li C, Zheng X, Zhang B, Wang H, Tao G, Xu J, Zhang Y, Han B. OCT4&SOX2-specific cytotoxic T lymphocytes plus programmed cell death protein 1 inhibitor presented with synergistic effect on killing lung cancer stem-like cells in vitro and treating drug-resistant lung cancer mice in vivo. J Cell Physiol 2018; 234:6758-6768. [PMID: 30382588 DOI: 10.1002/jcp.27423] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023]
Abstract
This study aimed to investigate the synergistic effect of octamer-binding transcription factor 4 and sex determining region Y-box 2 (OCT4&SOX2)-specific cytotoxic T lymphocytes (CTLs) and programmed cell death protein 1 (PD-1) inhibitor on killing lung cancer stem-like cells (LCSCs) and their efficacy in treating drug-resistant lung cancer (DRLC) mice. OCT4&SOX2-specific CTLs and PD-1 inhibitor with differed doses were applied to treat PC9 cells and PC9 LCSCs. Cell counting kit-8 (CCK8) assay and flow cytometry (FCM) assay with carboxyfluorescein diacetate/succinimidyl ester staining target cells before treatment and propidium iodide (PI) staining dead cells after treatment were conducted to detect the cytotoxic activity. DRLC mice were constructed by injection of PC9 LCSCs suspension and Matrigel into left lung of SD mice. DRLC mice were randomly divided into five groups: control group, cytomegalovirus (CMV) pp65 CTLs group, OCT4&SOX2 CTLs group, PD-1 inhibitor group, and OCT4&SOX2 CTLs + PD-1 inhibitor group. In vitro, both CCK8 assay and FCM assay disclosed that OCT4&SOX2-specific CTLs plus PD-1 inhibitor presented with elevated cytotoxic activity on PC9 cells and PC9 LCSCs. In vivo, tumor volume and tumor weight were decreased, while tumor necrosis and tumor apoptosis were increased in OCT4&SOX2 CTLs group than CMV pp65 CTLs group and control group, and in OCT4&SOX2 CTLs + PD-1 inhibitor group than OCT4&SOX2 CTLs group and PD-1 inhibitor group. In addition, CD8 expression was increased while OCT4&SOX2 expressions were decreased in OCT4&SOX2 CTLs + PD-1 inhibitor group than OCT4&SOX2 CTLs group and PD-1 inhibitor group. In conclusion, OCT4&SOX2-specific CTLs and PD-1 inhibitor presented with the synergistic effect on killing LCSCs in vitro and treating DRLC mice in vivo.
Collapse
Affiliation(s)
- Xueyan Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fang Hu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Changhui Li
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxuan Zheng
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huimin Wang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Guangyu Tao
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianlin Xu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanwei Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Baohui Han
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
44
|
Webb ES, Liu P, Baleeiro R, Lemoine NR, Yuan M, Wang Y. Immune checkpoint inhibitors in cancer therapy. J Biomed Res 2018; 32:317-326. [PMID: 28866656 PMCID: PMC6163118 DOI: 10.7555/jbr.31.20160168] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
In recent years immune checkpoint inhibitors have garnered attention as being one of the most promising types of immunotherapy on the horizon. There has been particular focus on the immune checkpoint molecules, cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1) which have been shown to have potent immunomodulatory effects through their function as negative regulators of T cell activation. CTLA-4, through engagement with its ligands B7-1 (CD80) and B7-2 (CD86), plays a pivotal role in attenuating the activation of naïve and memory T cells. In contrast, PD-1 is primarily involved in modulating T cell activity in peripheral tissues via its interaction with PD-L1 and PD-L2. The discovery of these negative regulators of the immune response was crucial in the development of checkpoint inhibitors. This shifted the focus from developing therapies that targeted activation of the host immune system against cancer to checkpoint inhibitors, which aimed to mediate tumor cell destruction through the removal of coinhibitory signals blocking anti-tumor T cell responses.
Collapse
Affiliation(s)
- Eika S. Webb
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
| | - Peng Liu
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
| | - Renato Baleeiro
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
| | - Nicholas R. Lemoine
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
- . Sino-British Research Centre for Molecular Oncology, National Center for International Research in Cell and Gene Therapy, Zhengzhou University, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450002, China
| | - Ming Yuan
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
| | - Yaohe Wang
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
- . Sino-British Research Centre for Molecular Oncology, National Center for International Research in Cell and Gene Therapy, Zhengzhou University, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450002, China
| |
Collapse
|
45
|
Hickey JW, Kosmides AK, Schneck JP. Engineering Platforms for T Cell Modulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:277-362. [PMID: 30262034 DOI: 10.1016/bs.ircmb.2018.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cells are crucial contributors to mounting an effective immune response and increasingly the focus of therapeutic interventions in cancer, infectious disease, and autoimmunity. Translation of current T cell immunotherapies has been hindered by off-target toxicities, limited efficacy, biological variability, and high costs. As T cell therapeutics continue to develop, the application of engineering concepts to control their delivery and presentation will be critical for their success. Here, we outline the engineer's toolbox and contextualize it with the biology of T cells. We focus on the design principles of T cell modulation platforms regarding size, shape, material, and ligand choice. Furthermore, we review how application of these design principles has already impacted T cell immunotherapies and our understanding of T cell biology. Recent, salient examples from protein engineering, synthetic particles, cellular and genetic engineering, and scaffolds and surfaces are provided to reinforce the importance of design considerations. Our aim is to provide a guide for immunologists, engineers, clinicians, and the pharmaceutical sector for the design of T cell-targeting platforms.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan P Schneck
- Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
46
|
Mimura K, Yamada L, Ujiie D, Hayase S, Tada T, Hanayama H, Thar Min AK, Shibata M, Momma T, Saze Z, Ohki S, Kono K. Immunotherapy for esophageal squamous cell carcinoma: a review. Fukushima J Med Sci 2018; 64:46-53. [PMID: 30058598 DOI: 10.5387/fms.2018-09] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer vaccines and immune checkpoint inhibitors (ICI) have recently been employed as immunotherapies for esophageal squamous cell carcinoma (ESCC). Cancer vaccines for ESCC have yielded several promising results from investigator-initiated phase I and II clinical trials. Furthermore, a Randomized Controlled Trial as an adjuvant setting after curative surgery is in progress in Japan. On the other hand, ICI, anti-CTLA-4 mAb and anti-PD-1 mAb, have demonstrated tumor shrinkage and improved overall survival in patients with multiple cancer types. For ESCC, several clinical trials using anti-PD-1/anti-PD-L1 mAb are underway with several recent promising results. In this review, cancer vaccines and ICI are discussed as novel therapeutic strategies for ESCC.
Collapse
Affiliation(s)
- Kosaku Mimura
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University.,Department of Advanced Cancer Immunotherapy, Fukushima Medical University.,Department of Progressive DOHaD Research, Fukushima Medical University
| | - Leo Yamada
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Daisuke Ujiie
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Suguru Hayase
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Takeshi Tada
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Hiroyuki Hanayama
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Aung Kyi Thar Min
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Masahiko Shibata
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University.,Department of Advanced Cancer Immunotherapy, Fukushima Medical University
| | - Tomoyuki Momma
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Zenichiro Saze
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Shinji Ohki
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| |
Collapse
|
47
|
Ma G, Deng Y, Jiang H, Li W, Wu Q, Zhou Q. The prognostic role of programmed cell death-ligand 1 expression in non-small cell lung cancer patients: An updated meta-analysis. Clin Chim Acta 2018; 482:101-107. [DOI: 10.1016/j.cca.2018.03.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 03/16/2018] [Accepted: 03/30/2018] [Indexed: 12/23/2022]
|
48
|
Zhang Q, Bi J, Zheng X, Chen Y, Wang H, Wu W, Wang Z, Wu Q, Peng H, Wei H, Sun R, Tian Z. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol 2018; 19:723-732. [PMID: 29915296 DOI: 10.1038/s41590-018-0132-0] [Citation(s) in RCA: 740] [Impact Index Per Article: 105.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 04/13/2018] [Indexed: 12/16/2022]
Abstract
Checkpoint blockade enhances effector T cell function and has elicited long-term remission in a subset of patients with a broad spectrum of cancers. TIGIT is a checkpoint receptor thought to be involved in mediating T cell exhaustion in tumors; however, the relevance of TIGIT to the dysfunction of natural killer (NK) cells remains poorly understood. Here we found that TIGIT, but not the other checkpoint molecules CTLA-4 and PD-1, was associated with NK cell exhaustion in tumor-bearing mice and patients with colon cancer. Blockade of TIGIT prevented NK cell exhaustion and promoted NK cell-dependent tumor immunity in several tumor-bearing mouse models. Furthermore, blockade of TIGIT resulted in potent tumor-specific T cell immunity in an NK cell-dependent manner, enhanced therapy with antibody to the PD-1 ligand PD-L1 and sustained memory immunity in tumor re-challenge models. This work demonstrates that TIGIT constitutes a previously unappreciated checkpoint in NK cells and that targeting TIGIT alone or in combination with other checkpoint receptors is a promising anti-cancer therapeutic strategy.
Collapse
Affiliation(s)
- Qing Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China.,Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Jiacheng Bi
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.,Shenzhen Laboratory of Fully Humanized Antibody Engineering, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaodong Zheng
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yongyan Chen
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Hua Wang
- the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenyong Wu
- the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhengguang Wang
- the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qiang Wu
- the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hui Peng
- Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiming Wei
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China.,Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Sun
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China. .,Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.
| | - Zhigang Tian
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China. .,Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
49
|
Update on Tumor Neoantigens and Their Utility: Why It Is Good to Be Different. Trends Immunol 2018; 39:536-548. [PMID: 29751996 DOI: 10.1016/j.it.2018.04.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/18/2022]
Abstract
Antitumor rejection by the immune system is a complex process that is regulated by several factors. Among these factors are the quality and quantity of mutational events that occur in cancer cells. Perhaps one of the most important types of mutations that influence antitumor immunity is the neoantigen, that is, a non-self-antigen that arises as a result of somatic mutation. Recent work has demonstrated that neoantigens hold significant promise for developing new diagnostic and therapeutic modalities. Therapeutic targeting of neoantigens is important for achieving benefit following therapy with immune checkpoint blockade agents or for cancer vaccines targeting mutations. Here, we review our understanding of neoantigens and discuss new developments in the quest to use them in cancer immunotherapy.
Collapse
|
50
|
Grenier JM, Yeung ST, Khanna KM. Combination Immunotherapy: Taking Cancer Vaccines to the Next Level. Front Immunol 2018; 9:610. [PMID: 29623082 PMCID: PMC5874308 DOI: 10.3389/fimmu.2018.00610] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/12/2018] [Indexed: 12/14/2022] Open
Abstract
With the advent of checkpoint blockade therapies, immunotherapy is now a critical modality for the treatment of some cancers. While some patients respond well to checkpoint blockade, many do not, necessitating the need for other forms of therapy. Vaccination against malignancy has been a long sought goal of science. For cancers holding a microbial etiology, vaccination has been highly effective in reducing the incidence of disease. However, vaccination against established malignancy has been largely disappointing. In this review, we discuss efforts to develop diverse vaccine modalities in the treatment of cancer with a particular focus on melanoma. Recent work has suggested that vaccines targeting patient-specific tumor mutations may be more relevant than those targeting unmutated proteins. Nonetheless, tumor cells utilize many strategies to evade host immunity. It is likely that the full potential of cancer vaccination will only be realized when vaccines are combined with other therapies targeting tumor immunoevasive mechanisms. By modulating inhibitory molecules, regulatory immune cells, and the metabolic resources and demands of T cells, scientists and clinicians can ensure vaccine-stimulated T cells are fully functional within the immunosuppressive tumor microevironment.
Collapse
Affiliation(s)
- Jeremy M Grenier
- Department of Immunology, University of Connecticut Health, Farmington, CT, United States
| | - Stephen T Yeung
- Department of Microbiology, New York University Langone School of Medicine, New York, NY, United States
| | - Kamal M Khanna
- Department of Immunology, University of Connecticut Health, Farmington, CT, United States.,Department of Microbiology, New York University Langone School of Medicine, New York, NY, United States.,Perlmutter Cancer Center, New York University Langone Health, New York, NY, United States
| |
Collapse
|