1
|
Giram P, Md Mahabubur Rahman K, Aqel O, You Y. In Situ Cancer Vaccines: Redefining Immune Activation in the Tumor Microenvironment. ACS Biomater Sci Eng 2025; 11:2550-2583. [PMID: 40223683 DOI: 10.1021/acsbiomaterials.5c00121] [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] [Indexed: 04/15/2025]
Abstract
Cancer is one of the leading causes of mortality worldwide. Nanomedicines have significantly improved life expectancy and survival rates for cancer patients in current standard care. However, recurrence of cancer due to metastasis remains a significant challenge. Vaccines can provide long-term protection and are ideal for preventing bacterial and viral infections. Cancer vaccines, however, have shown limited therapeutic efficacy and raised safety concerns despite extensive research. Cancer vaccines target and stimulate responses against tumor-specific antigens and have demonstrated great potential for cancer treatment in preclinical studies. However, tumor-associated immunosuppression and immune tolerance driven by immunoediting pose significant challenges for vaccine design. In situ vaccination represents an alternative approach to traditional cancer vaccines. This strategy involves the intratumoral administration of immunostimulants to modulate the growth and differentiation of innate immune cells, such as dendritic cells, macrophages, and neutrophils, and restore T-cell activity. Currently approved in situ vaccines, such as T-VEC, have demonstrated clinical promise, while ongoing clinical trials continue to explore novel strategies for broader efficacy. Despite these advancements, failures in vaccine research highlight the need to address tumor-associated immune suppression and immune escape mechanisms. In situ vaccination strategies combine innate and adaptive immune stimulation, leveraging tumor-associated antigens to activate dendritic cells and cross-prime CD8+ T cells. Various vaccine modalities, such as nucleotide-based vaccines (e.g., RNA and DNA vaccines), peptide-based vaccines, and cell-based vaccines (including dendritic, T-cell, and B-cell approaches), show significant potential. Plant-based viral approaches, including cowpea mosaic virus and Newcastle disease virus, further expand the toolkit for in situ vaccination. Therapeutic modalities such as chemotherapy, radiation, photodynamic therapy, photothermal therapy, and Checkpoint blockade inhibitors contribute to enhanced antigen presentation and immune activation. Adjuvants like CpG-ODN and PRR agonists further enhance immune modulation and vaccine efficacy. The advantages of in situ vaccination include patient specificity, personalization, minimized antigen immune escape, and reduced logistical costs. However, significant barriers such as tumor heterogeneity, immune evasion, and logistical challenges remain. This review explores strategies for developing potent cancer vaccines, examines ongoing clinical trials, evaluates immune stimulation methods, and discusses prospects for advancing in situ cancer vaccination.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kazi Md Mahabubur Rahman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Osama Aqel
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| |
Collapse
|
2
|
Giram P, Bist G, Woo S, Wohlfert E, Pili R, You Y. Prodrugs of paclitaxel improve in situ photo-vaccination. Photochem Photobiol 2024:10.1111/php.14025. [PMID: 39384406 PMCID: PMC11978925 DOI: 10.1111/php.14025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/25/2024] [Accepted: 08/22/2024] [Indexed: 10/11/2024]
Abstract
Photodynamic therapy (PDT) effectively kills cancer cells and initiates immune responses that promote anticancer effects locally and systemically. Primarily developed for local and regional cancers, the potential of PDT for systemic antitumor effects [in situ photo-vaccination (ISPV)] remains underexplored. This study investigates: (1) the comparative effectiveness of paclitaxel (PTX) prodrug [Pc-(L-PTX)2] for PDT and site-specific PTX effects versus its pseudo-prodrug [Pc-(NCL-PTX)2] for PDT combined with checkpoint inhibitors; (2) mechanisms driving systemic antitumor effects; and (3) the prophylactic impact on preventing cancer recurrence. A bilateral tumor model was established in BALB/c mice through subcutaneous injection of CT26 cells. Mice received the PTX prodrug (0.5 μmole kg-1, i.v.), and tumors were treated with a 690-nm laser (75 mW cm-2 for 30 min, drug-light interval 0.5 h, light does 135 J cm-1), followed by anti-CTLA-4 (100 μg dose-1, i.p.) on days 1, 4, and 7. Notable enhancement in both local and systemic antitumor effectiveness was observed with [Pc-(L-PTX)2] compared to [Pc-(NCL-PTX)2] with checkpoint inhibitor. Immune cell depletion and immunohistochemistry confirmed neutrophils and CD8+ T cells are effectors for systemic antitumor effects. Treatment-induced immune memory resisted newly rechallenged CT26, showcasing prophylactic benefits. ISPV with a PTX prodrug and anti-CTLA-4 is a promising approach for treating metastatic cancers and preventing recurrence.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Ganesh Bist
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Elizabeth Wohlfert
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY,14203, USA
| | - Roberto Pili
- Division of Hematology and Oncology, Department of Medicine, University at Buffalo, Buffalo, NY, 14203, USA
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| |
Collapse
|
3
|
Saku S, Toh U, Takao Y, Sakurai S, Yamada A, Shichijo S, Itoh K, Akagi Y. Plasma level of prostate related-antigen peptide-reactive IgG is a prognostic factor of patients with breast cancer treated with personalized peptide vaccines. Exp Ther Med 2021; 22:905. [PMID: 34249152 PMCID: PMC8264823 DOI: 10.3892/etm.2021.10337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 08/06/2020] [Indexed: 12/02/2022] Open
Abstract
The present study assessed plasma IgG in patients with metastatic recurrent breast cancer (mrBC) that is reactive to various T-cell epitope peptides of prostate-related antigens (PRAs), such as prostate-specific antigen, prostate-specific membrane antigen and prostate acid phosphatase. Patients were treated with personalized peptide vaccines (PPVs) which were selected and administered from a panel of candidate peptides based on human leukocyte antigen-types and prevaccination IgG levels to each peptide. The peptide panel consisted of 27 cytotoxic T-lymphocyte-epitope peptides derived from tumor-associated antigens, not including PRA. PRA peptides and peptide panels were retrospectively analyzed in 77 PPV-treated patients. The results revealed that PRA reactive IgG levels were increased after vaccination in 31 of the 97 patients included in the present study. Although there was no significant association between anti-PRA peptide levels and progression-free survival (PFS) or overall survival, anti-PRA peptide levels were significantly associated with PFS (P=0.009) in estrogen-receptor positive (ER+) patients with cancer. The results suggested that plasma anti-PRA IgG levels may be a useful prognostic marker for monitoring PPVs, particularly for ER+ patients with mrBC (trial registration no. from the UMIN Clinical Trials Registry, UMIN000001844).
Collapse
Affiliation(s)
- Shuko Saku
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Uhi Toh
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Yuko Takao
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Sayaka Sakurai
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Akira Yamada
- Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Shigeki Shichijo
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Yoshito Akagi
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
4
|
Amiri A, Pourhanifeh MH, Mirzaei HR, Nahand JS, Moghoofei M, Sahebnasagh R, Mirzaei H, Hamblin MR. Exosomes and Lung Cancer: Roles in Pathophysiology, Diagnosis and Therapeutic Applications. Curr Med Chem 2021; 28:308-328. [PMID: 32013817 DOI: 10.2174/0929867327666200204141952] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/18/2019] [Accepted: 11/29/2019] [Indexed: 11/22/2022]
Abstract
Lung cancer is a malignancy with a high morbidity and mortality rate, and affected patients have low survival and poor prognosis. The therapeutic approaches for the treatment of this cancer, including radiotherapy and chemotherapy, are not particularly effective partly due to late diagnosis. Therefore, the search for new diagnostic and prognostic tools is a critical issue. Novel biomarkers, such as exosomes, could be considered as potential diagnostic tools for malignancies, particularly lung cancer. Exosomes are nanovesicles, which are associated with different physiological and pathological conditions. It has been shown that these particles are released from many cells, such as cancer cells, immune cells and to some degree normal cells. Exosomes could alter the behavior of target cells through intercellular transfer of their cargo (e.g. DNA, mRNA, long non-coding RNAs, microRNAs and proteins). Thus, these vehicles may play pivotal roles in various physiological and pathological conditions. The current insights into lung cancer pathogenesis suggest that exosomes are key players in the pathogenesis of this cancer. Hence, these nanovesicles and their cargos could be used as new diagnostic, prognostic and therapeutic biomarkers in the treatment of lung cancer. Besides the diagnostic roles of exosomes, their use as drug delivery systems and as cancer vaccines is under investigation. The present review summarizes the current information on the diagnostic and pathogenic functions of exosomes in lung cancer.
Collapse
Affiliation(s)
- Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashahd, Iran
| | | | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, University of Medical Sciences, Tehran, Iran
| | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roxana Sahebnasagh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, United States
| |
Collapse
|
5
|
Noguchi M, Koga N, Moriya F, Suekane S, Yutani S, Yamada A, Shichijo S, Kakuma T, Itoh K. Survival analysis of multiple peptide vaccination for the selection of correlated peptides in urological cancers. Cancer Sci 2018; 109:2660-2669. [PMID: 29938870 PMCID: PMC6277968 DOI: 10.1111/cas.13709] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/16/2018] [Indexed: 12/24/2022] Open
Abstract
Peptide‐based cancer vaccines are able to induce strong immune responses, but their clinical results are unsatisfactory. To determine clinically correlated peptides, we analyzed survival data from urological cancer patients treated by personalized peptide vaccination (PPV), in which different multiple peptides were used for individual patients based on human leukocyte antigen (HLA) type and pre‐existing immunity. Survival data were obtained from a database of 265 urological cancer patients treated in 5 clinical PPV trials comprising 154 patients with castration‐resistant prostate cancer (CRPC) and 111 patients with advanced urothelial cancer (UC). Expression of tumor‐associated antigens (TAA) was evaluated in 10 prostate cancer tissues, 4 metastatic lymph nodes from prostate cancer, and 10 UC tissues using immunohistochemical staining. Clinical efficacy of individual peptides for overall survival was evaluated by the Cox proportional hazards regression model. All TAA coding candidate peptides used in PPV treatment were expressed in tumor cells from prostate cancer and UC samples except for p56Lck in both, and prostate‐specific antigen (PSA), prostatic acid phosphatase (PAP) and prostate‐specific membrane antigen (PSMA) in the UC samples. Patients with the following peptides had a significantly longer survival than patients without the peptides (hazard ratio <1.0, 95% confidence intervals <1.0 and P < .05): SART3‐109, PTHrP‐102, HNPRL‐140, SART3‐302 and Lck‐90 in CRPC patients, and EGF‐R‐800, Lck‐486, PSMA‐624, CypB‐129 and SART3‐734 in advanced UC patients, respectively. Correlated peptides selected using both survival data and pre‐existing immunity for PPV treatment may enhance the clinical benefits for urological cancer patients.
Collapse
Affiliation(s)
- Masanori Noguchi
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Shigeru Yutani
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Akira Yamada
- Division of Cancer Vaccines in Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Shigeki Shichijo
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Tatuyuki Kakuma
- Bio-statistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
6
|
Jung CY, Antonia SJ. Tumor Immunology and Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer. Tuberc Respir Dis (Seoul) 2018; 81:29-41. [PMID: 29332322 PMCID: PMC5771744 DOI: 10.4046/trd.2017.0120] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 11/13/2017] [Accepted: 11/26/2017] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is one of the most commonly diagnosed cancers and the leading cause of cancer-related deaths worldwide. Although progress in the treatment of advanced non-small cell lung cancer (NSCLC) has been made over the past decade, the 5-year survival rate in patients with lung cancer remains only 10%–20%. Obviously, new therapeutic options are required for patients with advanced NSCLC and unmet medical needs. Cancer immunotherapy is an evolving treatment modality that uses a patient's own immune systems to fight cancer. Theoretically, cancer immunotherapy can result in long-term cancer remission and may not cause the same side effects as chemotherapy and radiation. Immuno-oncology has become an important focus of basic research as well as clinical trials for the treatment of NSCLC. Immune checkpoint inhibitors are the most promising approach for cancer immunotherapy and they have become the standard of care for patients with advanced NSCLC. This review summarizes basic tumor immunology and the relevant clinical data on immunotherapeutic approaches, especially immune checkpoint inhibitors in NSCLC.
Collapse
Affiliation(s)
- Chi Young Jung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Scott J Antonia
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
7
|
Precision medicine for urothelial bladder cancer: update on tumour genomics and immunotherapy. Nat Rev Urol 2017; 15:92-111. [PMID: 29133939 DOI: 10.1038/nrurol.2017.179] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Effective management of advanced urothelial bladder cancer is challenging. New discoveries that improve our understanding of molecular bladder cancer subtypes have revealed numerous potentially targetable genomic alterations and demonstrated the efficacy of treatments that harness the immune system. These findings have begun to change paradigms of bladder cancer therapy. For example, DNA repair pathway mutations in genes such as ERCC2, FANCC, ATM, RB1, and others can predict responses to neoadjuvant platinum-based chemotherapies and to targeted therapies on the basis of mutation status. Furthermore, an increasing number of pan-cancer clinical trials (commonly referred to as basket or umbrella trials) are enrolling patients on the basis of molecular and genetic predictors of response. These studies promise to provide improved insight into the true utility of personalized medicine in the treatment of bladder cancer and many other cancer types. Finally, therapies that modulate immune responses have shown great benefit in many cancer types. Several immune checkpoint inhibitors that target programmed cell death protein 1 (PD1), its ligand PDL1, and cytotoxic T lymphocyte-associated protein 4 (CTLA4) have already been approved for use in bladder cancer, representing the most important change to the urological oncologist's tool-kit in over a decade. These advances also provide opportunities for personalization of bladder cancer therapy.
Collapse
|
8
|
Sakamoto S, Yoshitomi M, Yutani S, Terazaki Y, Yoshiyama K, Ioji T, Matsueda S, Yamada A, Takamori S, Itoh K, Hattori N, Kohno N, Sasada T. Evaluation of prognostic significance of granulocyte-related factors in cancer patients undergoing personalized peptide vaccination. Hum Vaccin Immunother 2016; 11:2784-9. [PMID: 26325075 PMCID: PMC5054776 DOI: 10.1080/21645515.2015.1075107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Since cancer vaccines do not always elicit beneficial effects in treated patients, identification of biomarkers for predicting clinical outcomes would be highly desirable. We previously reported that abnormal granulocytes present in peripheral blood mononuclear cells (PBMC) may contribute to poor prognosis in advanced prostate cancer patients receiving personalized peptide vaccination (PPV). In the current study, we examined whether soluble factors derived from granulocytes, such as matrix metalloproteinase 9 (MMP-9), myeloperoxidase (MPO), and arginase 1 (ARG1), and inhibitory cytokine TGFβ in pre-vaccination plasma were useful for predicting prognosis after PPV in advanced cancer patients. In biliary tract cancer (n=25), multivariate Cox regression analysis demonstrated that patients with higher plasma MMP-9 levels had a significantly worse overall survival (OS) [hazard ratio (HR) = 4.637, 95% confidence interval (CI) = 1.670 - 12.877, P = 0.003], whereas MPO, ARG1, or TGFβ levels were not correlated with OS. Similarly, patients with higher MMP-9 levels showed worse prognosis than those with lower MMP-9 levels in other types of advanced cancers, including non-small cell lung cancer (n=32, P = 0.037 by log-rank test), and pancreatic cancer (n=41, P = 0.042 by log-rank test). Taken together, plasma MMP-9 levels before vaccination might be potentially useful as a biomarker for selecting advanced cancer patients who would benefit from PPV.
Collapse
Affiliation(s)
- Shinjiro Sakamoto
- a Research Center for Innovative Cancer Therapy; Kurume University ; Kurume , Japan.,b Cancer Vaccine Center; Kurume University ; Kurume , Japan.,c Department of Molecular and Internal Medicine ; Institute of Biomedical & Health Sciences; Hiroshima University ; Hiroshima , Japan
| | - Munehiro Yoshitomi
- d Department of Surgery ; Kurume University School of Medicine ; Kurume , Japan
| | - Shigeru Yutani
- b Cancer Vaccine Center; Kurume University ; Kurume , Japan
| | - Yasuhiro Terazaki
- d Department of Surgery ; Kurume University School of Medicine ; Kurume , Japan
| | - Koichi Yoshiyama
- d Department of Surgery ; Kurume University School of Medicine ; Kurume , Japan
| | - Tetsuya Ioji
- b Cancer Vaccine Center; Kurume University ; Kurume , Japan
| | | | - Akira Yamada
- a Research Center for Innovative Cancer Therapy; Kurume University ; Kurume , Japan
| | - Shinzo Takamori
- d Department of Surgery ; Kurume University School of Medicine ; Kurume , Japan
| | - Kyogo Itoh
- b Cancer Vaccine Center; Kurume University ; Kurume , Japan
| | - Noboru Hattori
- c Department of Molecular and Internal Medicine ; Institute of Biomedical & Health Sciences; Hiroshima University ; Hiroshima , Japan
| | - Nobuoki Kohno
- c Department of Molecular and Internal Medicine ; Institute of Biomedical & Health Sciences; Hiroshima University ; Hiroshima , Japan
| | - Tetsuro Sasada
- b Cancer Vaccine Center; Kurume University ; Kurume , Japan.,e Kanagawa Cancer Center Research Institute ; Yokohama , Japan
| |
Collapse
|
9
|
Abstract
Cancer is one of the major leading death causes of diseases. Prevention and treatment of cancer is an important way to decrease the incidence of tumorigenesis and prolong patients' lives. Subversive achievements on cancer immunotherapy have recently been paid much attention after many failures in basic and clinical researches. Based on deep analysis of genomics and proteomics of tumor antigens, a variety of cancer vaccines targeting tumor antigens have been tested in preclinical and human clinical trials. Many therapeutic cancer vaccines alone or combination with other conventional treatments for cancer obtained spectacular efficacy, indicating the tremendously potential application in clinic. With the illustration of underlying mechanisms of cancer immune regulation, valid, controllable, and persistent cancer vaccines will play important roles in cancer treatment, survival extension and relapse and cancer prevention. This chapter mainly summarizes the recent progresses and developments on cancer vaccine research and clinical application, thus exploring the existing obstacles in cancer vaccine research and promoting the efficacy of cancer vaccine.
Collapse
|
10
|
Sakamoto S, Noguchi M, Yamada A, Itoh K, Sasada T. Prospect and progress of personalized peptide vaccinations for advanced cancers. Expert Opin Biol Ther 2016; 16:689-98. [PMID: 26938083 DOI: 10.1517/14712598.2016.1161752] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The field of cancer immunotherapy has made dramatic progress in the past 20 years, in part due to the identification of numerous tumor-associated antigens (TAAs). We have developed a novel immunotherapeutic approach called the personalized peptide vaccine (PPV), in which a maximum of four human leukocyte antigen (HLA)-matched vaccine peptides are selected based on the pre-existing host immunity before vaccination. AREAS COVERED This review describes recent progress in the use of PPV for various types of advanced cancer. EXPERT OPINION Although various approaches for therapeutic cancer immunotherapies, including peptide-based vaccines, have been developed and clinically examined, the diverse and heterogeneous characteristics of tumor cells and host immunity seem to limit their therapeutic efficacy. Selection of suitable peptide vaccines for individual patients based on the pre-existing host immunity before vaccination could resolve this limitation and could be a rational approach for developing effective cancer vaccines.
Collapse
Affiliation(s)
- Shinjiro Sakamoto
- a Research Center for Innovative Cancer Therapy , Kurume University , Kurume , Japan.,b Cancer Vaccine Center , Kurume University , Kurume , Japan.,c Department of Molecular and Internal Medicine School of Medicine, Graduate School of Biomedical and Health Sciences , Hiroshima University , Hiroshima , Japan
| | - Masanori Noguchi
- a Research Center for Innovative Cancer Therapy , Kurume University , Kurume , Japan
| | - Akira Yamada
- a Research Center for Innovative Cancer Therapy , Kurume University , Kurume , Japan
| | - Kyogo Itoh
- b Cancer Vaccine Center , Kurume University , Kurume , Japan
| | - Tetsuro Sasada
- b Cancer Vaccine Center , Kurume University , Kurume , Japan.,d Kanagawa Cancer Center Research Institute , Yokohama , Japan
| |
Collapse
|
11
|
Seledtsov VI, Goncharov AG, Seledtsova GV. Clinically feasible approaches to potentiating cancer cell-based immunotherapies. Hum Vaccin Immunother 2016; 11:851-69. [PMID: 25933181 DOI: 10.1080/21645515.2015.1009814] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The immune system exerts both tumor-destructive and tumor-protective functions. Mature dendritic cells (DCs), classically activated macrophages (M1), granulocytes, B lymphocytes, aβ and ɣδ T lymphocytes, natural killer T (NKT) cells, and natural killer (NK) cells may be implicated in antitumor immunoprotection. Conversely, tolerogenic DCs, alternatively activated macrophages (M2), myeloid-derived suppressor cells (MDSCs), and regulatory T (Tregs) and B cells (Bregs) are capable of suppressing antitumor immune responses. Anti-cancer vaccination is a useful strategy to elicit antitumor immune responses, while overcoming immunosuppressive mechanisms. Whole tumor cells or lysates derived thereof hold more promise as cancer vaccines than individual tumor-associated antigens (TAAs), because vaccinal cells can elicit immune responses to multiple TAAs. Cancer cell-based vaccines can be autologous, allogeneic or xenogeneic. Clinical use of xenogeneic vaccines is advantageous in that they can be most effective in breaking the preexisting immune tolerance to TAAs. To potentiate immunotherapy, vaccinations can be combined with other modalities that target different immune pathways. These modalities include 1) genetic or chemical modification of cell-based vaccines; 2) cross-priming TAAs to T cells by engaging dendritic cells; 3) T-cell adoptive therapy; 4) stimulation of cytotoxic inflammation by non-specific immunomodulators, toll-like receptor (TLR) agonists, cytokines, chemokines or hormones; 5) reduction of immunosuppression and/or stimulation of antitumor effector cells using antibodies, small molecules; and 6) various cytoreductive modalities. The authors envisage that combined immunotherapeutic strategies will allow for substantial improvements in clinical outcomes in the near future.
Collapse
Key Words
- ADCC, antibody-dependent cell cytotoxicity
- APC, antigen-presenting cell
- Ab, antibodies
- BCG, Bacillus Calmette-Guérin
- Breg, regulatory B cell
- CAR, chimeric antigen receptor
- COX, cyclooxygenase
- CTA, cancer/testis antigen
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T lymphocyte antigen-4
- DC, dendritic cell
- DTH, delayed-type hypersensitivity
- GITR, glucocorticoid-induced tumor necrosis factor receptor
- GM-CSF, granulocyte-macrophage colony stimulating factor
- HIFU, high-intensity focused ultrasound
- IDO, indoleamine-2, 3-dioxygenase
- IFN, interferon
- IL, interleukin
- LAK, lymphokine-activated killer
- M, macrophage
- M1, classically activated macrophage
- M2, alternatively activated macrophage, MDSC, myeloid-derived suppressor cell
- MHC, major histocompatibility complex
- NK, natural killer (cell)
- PD-1, programmed death-1
- PGE2, prostaglandin E2
- RFA, radiofrequency ablation
- RNS, reactive nitrogen species
- ROS
- TAA, tumor-associated antigen
- TGF, transforming growth factor
- TLR, toll-like receptor
- TNF, tumor necrosis factor
- Th, T-helper cell
- Treg, regulatory T cell
- VEGF, vascular endothelial growth factor
- antitumor immunoprotection
- cancer cell-based vaccines
- combined immunotherapy
- immunosuppression
- reactive oxygen species
Collapse
Affiliation(s)
- V I Seledtsov
- a lmmanuel Kant Baltic Federal University ; Kaliningrad , Russia
| | | | | |
Collapse
|
12
|
Noguchi M, Moriya F, Koga N, Matsueda S, Sasada T, Yamada A, Kakuma T, Itoh K. A randomized phase II clinical trial of personalized peptide vaccination with metronomic low-dose cyclophosphamide in patients with metastatic castration-resistant prostate cancer. Cancer Immunol Immunother 2016; 65:151-60. [PMID: 26728480 PMCID: PMC11028889 DOI: 10.1007/s00262-015-1781-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/11/2015] [Indexed: 01/21/2023]
Abstract
This study investigated the effect of metronomic cyclophosphamide (CPA) in combination with personalized peptide vaccination (PPV) on regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC), and whether it could improve the antitumor effect of PPV. Seventy patients with metastatic castration-resistant prostate cancer were randomly assigned (1:1) to receive PPV plus oral low-dose CPA (50 mg/day), or PPV alone. PPV treatment used a maximum of four peptides chosen from 31 pooled peptides according to human leukocyte antigen types and antigen-specific humoral immune responses before PPV, for 8 subcutaneous weekly injections. Peptide-specific cytotoxic T lymphocyte (CTL) and immunoglobulin G responses were measured before and after PPV. The incidence of grade 3 or 4 hematologic adverse events was higher in the PPV plus CPA arm than in the PPV alone arm. Decrease in Treg and increase in MDSC were more pronounced in PPV plus CPA treatment than in PPV alone (p = 0.036 and p = 0.048, respectively). There was no correlation between the changes in Treg or MDSC and CTL response. There was no difference in positive immune responses between the two arms, although overall survival in patients with positive immune responses was longer than in those with negative immune responses (p = 0.001). Significant differences in neither progression-free survival nor overall survival were observed between the two arms. Low-dose CPA showed no change in the antitumor effect of PPV, possibly due to the simultaneous decrease in Treg and increase in MDSC, in patients under PPV.
Collapse
MESH Headings
- Administration, Metronomic
- Aged
- Aged, 80 and over
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/immunology
- Combined Modality Therapy
- Cyclophosphamide/administration & dosage
- Humans
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasm Metastasis
- Peptides/administration & dosage
- Peptides/chemistry
- Peptides/immunology
- Precision Medicine/methods
- Prostatic Neoplasms, Castration-Resistant/immunology
- Prostatic Neoplasms, Castration-Resistant/mortality
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/therapy
- Survival Analysis
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Treatment Outcome
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/adverse effects
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Masanori Noguchi
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan.
- Department of Urology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan.
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan.
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Satoko Matsueda
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Tetsuro Sasada
- Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| | - Akira Yamada
- Division of Cancer Vaccines, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuyuki Kakuma
- Bio-statistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
13
|
Noguchi M, Arai G, Matsumoto K, Naito S, Moriya F, Suekane S, Komatsu N, Matsueda S, Sasada T, Yamada A, Kakuma T, Itoh K. Phase I trial of a cancer vaccine consisting of 20 mixed peptides in patients with castration-resistant prostate cancer: dose-related immune boosting and suppression. Cancer Immunol Immunother 2015; 64:493-505. [PMID: 25662406 PMCID: PMC11028456 DOI: 10.1007/s00262-015-1660-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 01/16/2015] [Indexed: 12/11/2022]
Abstract
The heterogeneity expression of tumor-associated antigens (TAA) and variability of human T cell repertoire suggest that effective cancer vaccine requires induction of a wide breadth of cytotoxic T lymphocyte (CTL) specificities. This can be achieved with vaccines targeting multiple TAA. We evaluated the safety and immune dynamics of a cancer vaccine consisting of 20 mixed peptides (KRM-20) designed to induce CTLs against 12 different TAA in patients with castration-resistant prostate cancer (CRPC). Patients received each of three different randomly assigned doses of KRM-20 (6, 20, or 60 mg) once a week for 6 weeks. KRM-20 was applicable for patients with positive human leukocyte antigen (HLA) A2, A3, A11, A24, A26, A31 or A33 alleles, which cover the majority of the global population. To evaluate the minimum immunological effective dose (MIED), peptide-specific CTL and immunoglobulin G (IgG) responses, and immune suppressive subsets were evaluated during the vaccination. Total of 17 patients was enrolled. No serious adverse drug reactions were encountered. The MIED of KRM-20 in CTL or IgG response calculated by logistic regression model was set as 16 or 1.6 mg, respectively. The frequency of immune suppressive subsets was fewer in the 20 mg cohort than that in 6 or 60 mg cohort. Clinical responses determined by prostate-specific antigen levels were two partial responses (from the 20 mg cohort), five no changes and ten progressive diseases. Twenty milligrams of KRM-20 could be recommended for further studies because of the safety and ability to augment CTL activity.
Collapse
Affiliation(s)
- Masanori Noguchi
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Barbosa T, Barral-Netto M. Challenges in the research and development of new human vaccines. Braz J Med Biol Res 2015; 46:103-8. [PMID: 23558931 PMCID: PMC3854358 DOI: 10.1590/1414-431x20131873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 01/16/2013] [Indexed: 12/22/2022] Open
Abstract
The field of vaccinology was born from the observations by the fathers of vaccination, Edward Jenner and Louis Pasteur, that a permanent, positive change in the way our bodies respond to life-threatening infectious diseases can be obtained by specific challenge with the inactivated infectious agent performed in a controlled manner, avoiding the development of clinical disease upon exposure to the virulent pathogen. Many of the vaccines still in use today were developed on an empirical basis, essentially following the paradigm established by Pasteur, "isolate, inactivate, and inject" the disease-causing microorganism, and are capable of eliciting uniform, long-term immune memory responses that constitute the key to their proven efficacy. However, vaccines for pathogens considered as priority targets of public health concern are still lacking. The literature tends to focus more often on vaccine research problems associated with specific pathogens, but it is increasingly clear that there are common bottlenecks in vaccine research, which need to be solved in order to advance the development of the field as a whole. As part of a group of articles, the objective of the present report is to pinpoint these bottlenecks, exploring the literature for common problems and solutions in vaccine research applied to different situations. Our goal is to stimulate brainstorming among specialists of different fields related to vaccine research and development. Here, we briefly summarize the topics we intend to deal with in this discussion.
Collapse
Affiliation(s)
- T Barbosa
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz, Salvador, BA, Brasil.
| | | |
Collapse
|
15
|
Feasibility study of personalized peptide vaccination for metastatic recurrent triple-negative breast cancer patients. Breast Cancer Res 2014; 16:R70. [PMID: 24992895 PMCID: PMC4227005 DOI: 10.1186/bcr3685] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 06/23/2014] [Indexed: 12/15/2022] Open
Abstract
Introduction Since treatment modalities for metastatic recurrent triple-negative breast cancer (mrTNBC) are limited, a novel treatment approach including immunotherapy is required. We have developed a novel regimen of personalized peptide vaccination (PPV), in which vaccine antigens are individually selected from a pool of different peptide candidates based on the pre-existing host immunity. Herein we conducted a phase II study of PPV for metastatic recurrent breast cancer patients to investigate the feasibility of PPV for mrTNBC. Methods Seventy-nine patients with metastatic recurrent breast cancer who had metastases and had failed standard chemotherapy and/or hormonal therapy were enrolled. They were subgrouped as the mrTNBC group (n = 18), the luminal/human epidermal growth factor receptor 2 (HER2)-negative group (n = 41) and the HER2-positive group (n = 18), while the remaining two patients had not been investigated. A maximum of four human leukocyte antigen (HLA)-matched peptides showing higher peptide-specific immunoglobulin G (IgG) responses in pre-vaccination plasma were selected from 31 pooled peptide candidates applicable for the four HLA-IA phenotypes (HLA-A2, -A24, or -A26 types, or HLA-A3 supertypes), and were subcutaneously administered weekly for 6 weeks and bi-weekly thereafter. Measurement of peptide-specific cytotoxic T lymphocyte (CTL) and IgG responses along with other laboratory analyses were conducted before and after vaccination. Results No severe adverse events associated with PPV were observed in any of the enrolled patients. Boosting of CTL and/or IgG responses was observed in most of the patients after vaccination, irrespective of the breast cancer subtypes. There were three complete response cases (1 mrTNBC and 2 luminal/HER2-negative types) and six partial response cases (1 mrTNBC and 5 luminal/HER2-negative types). The median progression-free survival time and median overall survival time of mrTNBC patients were 7.5 and 11.1 months, while those of luminal/HER2-negative patients were 12.2 and 26.5 months, and those of HER2-positive patients were 4.5 and 14.9 months, respectively. Conclusions PPV could be feasible for mrTNBC patients because of the safety, immune responses, and possible clinical benefits. Clinical Trial Registration Number UMIN000001844 (Registration Date: April 5, 2009)
Collapse
|
16
|
Azuma K, Komatsu N, Hattori S, Matsueda S, Kawahara A, Sasada T, Itoh K, Hoshino T. Humoral immune responses to EGFR-derived peptides predict progression-free and overall survival of non-small cell lung cancer patients receiving gefitinib. PLoS One 2014; 9:e86667. [PMID: 24497964 PMCID: PMC3909003 DOI: 10.1371/journal.pone.0086667] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 12/12/2013] [Indexed: 11/19/2022] Open
Abstract
Somatic mutations in the epidermal growth factor receptor (EGFR) gene are associated with clinical response to EGFR tyrosine kinase inhibitors (TKIs), such as gefitinib, in patients with non-small cell lung cancer (NSCLC). However, humoral immune responses to EGFR in NSCLC patients have not been well studied. In this study, we investigated the clinical significance of immunoglobulin G (IgG) responses to EGFR-derived peptides in NSCLC patients receiving gefitinib. Plasma IgG titers to each of 60 different EGFR-derived 20-mer peptides were measured by the Luminex system in 42 NSCLC patients receiving gefitinib therapy. The relationships between the peptide-specific IgG titers and presence of EGFR mutations or patient survival were evaluated statistically. IgG titers against the egfr_481–500, egfr_721–740, and egfr_741–760 peptides were significantly higher in patients with exon 21 mutation than in those without it. On the other hand, IgG titers against the egfr_841–860 and egfr_1001–1020 peptides were significantly lower and higher, respectively, in patients with deletion in exon 19. Multivariate Cox regression analysis showed that IgG responses to egfr_41_ 60, egfr_61_80 and egfr_481_500 were significantly prognostic for progression-free survival independent of other clinicopathological characteristics, whereas those to the egfr_41_60 and egfr_481_500 peptides were significantly prognostic for overall survival. Detection of IgG responses to EGFR-derived peptides may be a promising method for prognostication of NSCLC patients receiving gefitinib. Our results may provide new insight for better understanding of humoral responses to EGFR in NSCLC patients.
Collapse
Affiliation(s)
- Koichi Azuma
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
- * E-mail:
| | - Nobukazu Komatsu
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Satoshi Hattori
- Biostatistics Center, Kurume University, Kurume, Fukuoka, Japan
| | - Satoko Matsueda
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka, Japan
| | - Tetsuro Sasada
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kyogo Itoh
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| |
Collapse
|
17
|
Arlen PM, Wood LV. Prostate cancer vaccines: moving therapeutic vaccination forward in the post-Provenge™ era. Expert Rev Vaccines 2014; 11:287-302. [DOI: 10.1586/erv.11.183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
EGFR T790M mutation as a possible target for immunotherapy; identification of HLA-A*0201-restricted T cell epitopes derived from the EGFR T790M mutation. PLoS One 2013; 8:e78389. [PMID: 24223798 PMCID: PMC3818324 DOI: 10.1371/journal.pone.0078389] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/19/2013] [Indexed: 12/22/2022] Open
Abstract
Treatment with epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), such as gefitinib and erlotinib, has achieved high clinical response rates in patients with non–small cell lung cancers (NSCLCs). However, over time, most tumors develop acquired resistance to EGFR-TKIs, which is associated with the secondary EGFR T790M resistance mutation in about half the cases. Currently there are no effective treatment options for patients with this resistance mutation. Here we identified two novel HLA-A*0201 (A2)-restricted T cell epitopes containing the mutated methionine residue of the EGFR T790M mutation, T790M-5 (MQLMPFGCLL) and T790M-7 (LIMQLMPFGCL), as potential targets for EGFR-TKI-resistant patients. When peripheral blood cells were repeatedly stimulated in vitro with these two peptides and assessed by antigen-specific IFN-γ secretion, T cell lines responsive to T790M-5 and T790M-7 were established in 5 of 6 (83%) and 3 of 6 (50%) healthy donors, respectively. Additionally, the T790M-5- and T790M-7-specific T cell lines displayed an MHC class I-restricted reactivity against NSCLC cell lines expressing both HLA-A2 and the T790M mutation. Interestingly, the NSCLC patients with antigen-specific T cell responses to these epitopes showed a significantly less frequency of EGFR-T790M mutation than those without them [1 of 7 (14%) vs 9 of 15 (60%); chi-squared test, p = 0.0449], indicating the negative correlation between the immune responses to the EGFR-T790M-derived epitopes and the presence of EGFR-T790M mutation in NSCLC patients. This finding could possibly be explained by the hypothesis that immune responses to the mutated neo-antigens derived from T790M might prevent the emergence of tumor cell variants with the T790M resistance mutation in NSCLC patients during EGFR-TKI treatment. Together, our results suggest that the identified T cell epitopes might provide a novel immunotherapeutic approach for prevention and/or treatment of EGFR-TKI resistance with the secondary EGFR T790M resistance mutation in NSCLC patients.
Collapse
|
19
|
Takahashi R, Ishibashi Y, Hiraoka K, Matsueda S, Kawano K, Kawahara A, Kage M, Ohshima K, Yamanaka R, Shichijo S, Shirouzu K, Itoh K, Sasada T. Phase II study of personalized peptide vaccination for refractory bone and soft tissue sarcoma patients. Cancer Sci 2013; 104:1285-94. [PMID: 23829867 PMCID: PMC7656559 DOI: 10.1111/cas.12226] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 06/28/2013] [Accepted: 06/30/2013] [Indexed: 12/22/2022] Open
Abstract
Refractory bone and soft tissue sarcomas are challenging diseases to treat because of their robustness to chemotherapy. Although cancer vaccines have the potential to become an attractive treatment modality, their progress has been hampered by the presence of many subtypes of sarcomas and different human leukocyte antigen (HLA)‐types. We investigated whether personalized peptide vaccination (PPV) would be feasible for the vast majority of sarcoma patients. Twenty refractory bone and soft tissue sarcoma patients with nine different subtypes and 11 different HLA‐class IA phenotypes were enrolled in this study. A maximum of four HLA‐matched peptides showing higher peptide‐specific IgG responses in pre‐vaccination plasma were selected from 31 pooled peptide candidates applicable for the HLA‐A2, ‐A3, ‐A11, ‐A24, ‐A26, ‐A31, and ‐A33 types, and were subcutaneously administered weekly for 6 weeks and bi‐weekly thereafter. Measurement of peptide‐specific CTL and IgG responses along with other laboratory analyses were conducted before and after vaccination. No patients were excluded by either sarcoma subtypes or different HLA‐types. No severe adverse events associated with PPV were observed in any patients. Peptide‐specific immunological boosting was observed in the post‐vaccination samples from the majority of patients. Tumor reduction of the lung metastasis and a long stable disease was observed in each case, and the median overall survival time of the 20 cases was 9.6 months. Taken together, PPV could be feasible for the vast majority of refractory sarcoma patients because of the safety and higher rates of immunological responses regardless of the presence of different sarcoma subtypes and various HLA‐types.
Collapse
Affiliation(s)
- Ryuji Takahashi
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Denies S, Sanders NN. Recent progress in canine tumor vaccination: potential applications for human tumor vaccines. Expert Rev Vaccines 2013; 11:1375-86. [PMID: 23249236 DOI: 10.1586/erv.12.104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor vaccination holds great promise for the treatment of cancer and research concerning tumor vaccination in dogs is of great interest for veterinary as well as human medicine. Indeed, cancer is the leading cause of death in adult dogs and companion animals are acknowledged as excellent preclinical models for human oncology. The license of the veterinary melanoma vaccine (Oncept™) and Provenge® for the treatment of prostate cancer in men established tumor vaccination as a valid treatment modality for cancer. Although the results with this and other vaccines are promising, there are still some hurdles to overcome. In this article, preclinical and clinical trials with tumor vaccines in dogs are discussed, as well as the surplus value of canine cancer patients for human medicine.
Collapse
Affiliation(s)
- Sofie Denies
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | | |
Collapse
|
21
|
Noguchi M, Sasada T, Itoh K. Personalized peptide vaccination: a new approach for advanced cancer as therapeutic cancer vaccine. Cancer Immunol Immunother 2013; 62:919-29. [PMID: 23197273 PMCID: PMC11029009 DOI: 10.1007/s00262-012-1379-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 11/16/2012] [Indexed: 12/19/2022]
Abstract
Since both tumor cells and host immune cell repertoires are diverse and heterogeneous, immune responses against tumor-associated antigens should differ substantially among individual cancer patients. Selection of suitable peptide vaccines for individual patients based on the preexisting host immunity before vaccination could induce potent anti-tumor responses that provide clinical benefit to cancer patients. We have developed a novel immunotherapeutic approach of personalized peptide vaccination (PPV) in which a maximum of four human leukocyte antigen (HLA) class IA-matched peptides are selected for vaccination among pooled peptides on the basis of both HLA class IA type and the preexisting host immunity before vaccination. In this review, we discuss our recent results of preclinical and clinical studies of PPV for various types of advanced cancer.
Collapse
Affiliation(s)
- Masanori Noguchi
- Department of Urology, Kurume University School of Medicine, Kurume, Japan.
| | | | | |
Collapse
|
22
|
Tumor Inhibition by DepoVax-Based Cancer Vaccine Is Accompanied by Reduced Regulatory/Suppressor Cell Proliferation and Tumor Infiltration. ISRN ONCOLOGY 2013; 2013:753427. [PMID: 23533812 PMCID: PMC3606802 DOI: 10.1155/2013/753427] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 02/06/2013] [Indexed: 12/18/2022]
Abstract
A successful cancer vaccine needs to overcome the effects of immune-suppressor cells such as Treg lymphocytes, suppressive cytokine-secreting Tr1 cells, and myeloid-derived suppressor cells (MDSCs), while enhancing tumor-specific immune responses. Given the relative poor efficacy associated with current cancer vaccines, a novel vaccine platform called DepoVaxTM
(DPX) was developed. C3 tumor-challenged mice were immunized with HPV-E7 peptide in DPX- or conventional-emulsion- (CE-) based vaccine. While control mice showed marked increase in Treg/MDSCs in spleen and blood, in mice treated with DPX-E7 the levels remained similar to tumor-free naive mice. Such differences were also seen within the tumor. Antigen-specific IL10-secreting CD4/CD8 T cells and TGF-β+CD8+ T cell frequencies were increased significantly in CE-treated and control mice in contrast to DPX-E7-immunized mice. Analysis of tumor-infiltrating cells revealed higher frequency of suppressor cells in untreated controls than in DPX-E7 group while the converse was true for tumor-infiltrating CD8 T cells. Immunization of tumor-bearing HLA-A2 transgenic mice with human vaccine DPX-0907, a peptide-based vaccine for breast/ovarian/prostate cancers, showed efficient induction of immune response to cancer peptides despite the presence of suppressor cells. Thus, this study provides the rationale for using DPX-based cancer vaccines in immune-suppressed cancer patients, to induce effective anticancer immunity.
Collapse
|
23
|
Takahashi R, Yoshitomi M, Yutani S, Shirahama T, Noguchi M, Yamada A, Itoh K, Sasada T. Current status of immunotherapy for the treatment of biliary tract cancer. Hum Vaccin Immunother 2013; 9:1069-72. [PMID: 23376808 DOI: 10.4161/hv.23844] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biliary tract cancer (BTC) is one of the most aggressive malignancies. Although various promising regimens of chemotherapeutic and/or molecular targeted agents have been developed, further treatment modalities, including immunotherapies, still remain to be established for refractory patients who are unresponsive to or relapse after currently available therapeutic options for BTC. Recently, several clinical trials of immunotherapies, including peptide-based vaccines and dendritic cell (DC)-based vaccines, have been reported with promising results. Here we summarize the data from phase I or phase II clinical trials of immunotherapies for BTC. In particular, we introduce our novel immunotherapeutic approach called personalized peptide vaccine (PPV), in which HLA-matched peptides were selected and administered based on the pre-existing host immunity before vaccination, for the treatment of advanced BTC. Further clinical trials would be recommended to prove clinical benefits of these novel immunotherapeutic approaches. Recently concomitant treatments, such as chemotherapies and immune checkpoint blockade, have been reported to enhance the therapeutic effects of cancer immunotherapies through multiple coordinated immune mechanisms. Additional therapies in combination with immunotherapies could produce synergistic effects in the treatment of advanced BTC.
Collapse
Affiliation(s)
- Ryuji Takahashi
- Department of Surgery; Kurume University School of Medicine; Kurume, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Sasada T, Noguchi M, Yamada A, Itoh K. Personalized peptide vaccination: a novel immunotherapeutic approach for advanced cancer. Hum Vaccin Immunother 2012; 8:1309-13. [PMID: 22894962 DOI: 10.4161/hv.20988] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Since both tumor cells and immune cell repertoires are diverse and heterogeneous, immune responses against tumor-associated antigens might be substantially different among individual patients. Personalized selection of right peptides for individuals could thus be an appropriate strategy for cancer vaccines. We have developed a novel immunotherapeutic approach, personalized peptide vaccination (PPV), in which HLA-matched peptides are selected and administered, based on the pre-existing host immunity before vaccination. Recent clinical trials of PPV have demonstrated a feasibility of this new therapeutic approach in various types of advanced cancers. For example, a randomized phase II trial for patients with castration resistant prostate cancer showed a possible clinical benefit in the PPV group. In the patients undergoing PPV, lymphocyte counts, increased IgG responses to the vaccine peptides, and inflammatory factors in pre-vaccination peripheral blood might be potential biomarkers for prognosis. Further randomized phase III trials would be recommended to prove clinical benefits of PPV.
Collapse
Affiliation(s)
- Tetsuro Sasada
- Department of Immunology and Immunotherapy, Kurume University School of Medicine, Kurume, Japan.
| | | | | | | |
Collapse
|
25
|
Yoshitomi M, Yutani S, Matsueda S, Ioji T, Komatsu N, Shichijo S, Yamada A, Itoh K, Sasada T, Kinoshita H. Personalized peptide vaccination for advanced biliary tract cancer: IL-6, nutritional status and pre-existing antigen-specific immunity as possible biomarkers for patient prognosis. Exp Ther Med 2011; 3:463-469. [PMID: 22969912 DOI: 10.3892/etm.2011.424] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 11/22/2011] [Indexed: 12/31/2022] Open
Abstract
Considering that the prognosis of patients with advanced biliary tract cancer (BTC) remains very poor, with a median survival of less than 1 year, new therapeutic approaches need to be developed. In the present study, a phase II clinical trial of personalized peptide vaccination (PPV) was conducted in advanced BTC patients to evaluate the feasibility of this treatment and to identify potential biomarkers. A maximum of 4 human leukocyte antigen-matched peptides, which were selected based on the pre-existing host immunity prior to vaccination, were subcutaneously administered (weekly for 6 consecutive weeks and bi-weekly thereafter) to 25 advanced BTC patients without severe adverse events. Humoral and/or T cell responses specific to the vaccine antigens were substantially induced in a subset of the vaccinated patients. As shown by multivariate Cox regression analysis, lower interleukin-6 (IL-6) and higher albumin levels prior to vaccination and greater numbers of selected vaccine peptides were significantly favorable factors for overall survival [hazard ratio (HR)=1.123, 95% confidence interval (CI) 1.008-1.252, P=0.035; HR=0.158, 95% CI 0.029-0.860, P=0.033; HR=0.258, 95% CI 0.098-0.682, P=0.006; respectively]. Based on the safety profile and substantial immune responses to vaccine antigens, PPV could be a promising approach for refractory BTC, although its clinical efficacy remains to be investigated in larger-scale prospective studies. The identified biomarkers are potentially useful for selecting BTC patients who would benefit from PPV.
Collapse
|
26
|
Komatsu N, Matsueda S, Tashiro K, Ioji T, Shichijo S, Noguchi M, Yamada A, Doi A, Suekane S, Moriya F, Matsuoka K, Kuhara S, Itoh K, Sasada T. Gene expression profiles in peripheral blood as a biomarker in cancer patients receiving peptide vaccination. Cancer 2011; 118:3208-21. [PMID: 22071976 DOI: 10.1002/cncr.26636] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/26/2011] [Accepted: 09/16/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Because only a subset of patients show clinical responses to peptide-based cancer vaccination, it is critical to identify biomarkers for selecting patients who would most likely benefit from this treatment. METHODS The authors characterized the gene expression profiles in peripheral blood of vaccinated patients to identify biomarkers to predict patient prognosis. Peripheral blood was obtained from advanced castration-resistant prostate cancer patients, who survived for >900 days (long-term survivors, n = 20) or died within 300 days (short-term survivors, n = 20) after treatment with personalized peptide vaccination. Gene expression profiles in prevaccination and postvaccination peripheral blood mononuclear cells (PBMCs) were assessed by DNA microarray. RESULTS There were no statistically significant differences in the clinical or pathological features between the 2 groups. Microarray analysis of prevaccination PBMCs identified 19 genes that were differentially expressed between the short-term and long-term survivors. Among the 15 up-regulated genes in the short-term survivors, 13 genes, which were also differentially expressed in postvaccination PBMCs, were associated with gene signatures of granulocytes. When a set of 4 differentially expressed genes were selected as the best combination to determine patient survival, prognosis was correctly predicted in 12 of 13 patients in a validation set (accuracy, 92%). CONCLUSIONS These results suggested that abnormal granulocytes present in the PBMC faction may contribute to poor prognosis in advanced prostate cancer patients receiving personalized peptide vaccination. Gene expression profiling in peripheral blood might thus be informative for devising better therapeutic strategies by predicting patient prognosis after cancer vaccines.
Collapse
Affiliation(s)
- Nobukazu Komatsu
- Department of Immunology and Immunotherapy, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wu YD, Zhou GX. Recent advances in immunotherapy for human pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2011; 19:2091-2096. [DOI: 10.11569/wcjd.v19.i20.2091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the common highly malignant digestive system tumors. It is associated with a high mortality rate because of early metastasis, low resection rate and poor chemoradiotherapy response. Advances in immunology, such as tumor antigen, immunological surveillance, immunological evasion, immunological tolerance, T cell signal transduction, regulation of cytokines and regulatory DCs, down-regulation of co-stimulatory molecules, and tumor microenvironment, have enriched our knowledge and promoted the development of immunotherapy for pancreatic cancer. In this paper, we will review the recent advances in immunotherapy for pancreatic cancer.
Collapse
|
28
|
Matsumoto K, Noguchi M, Satoh T, Tabata KI, Fujita T, Iwamura M, Yamada A, Komatsu N, Baba S, Itoh K. A phase I study of personalized peptide vaccination for advanced urothelial carcinoma patients who failed treatment with methotrexate, vinblastine, adriamycin and cisplatin. BJU Int 2010; 108:831-8. [PMID: 21166757 DOI: 10.1111/j.1464-410x.2010.09933.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE • To investigate the safety and immune responses of 12 consecutive weeks of once-weekly personalized peptide vaccine (PPV) administration in patients with advanced urothelial carcinoma (UC) for whom therapy with methotrexate, vinblastine, adriamycin and cisplatin (MVAC) has failed. PATIENTS AND METHODS • A phase I trial was designed. Ten patients with MVAC-refractory advanced or metastatic UC were treated with weekly personalized peptide vaccine 12 times using positive peptides chosen from 14 and 16 peptides in patients with human leucocyte antigens A24 and A2, respectively. • Peptide-specific cytotoxic T lymphocyte precursor analysis by interferon-γ production and peptide-reactive immunoglobulin G (IgG) using an enzyme-linked immunosorbent assay was monitored during the treatment. RESULTS • The peptide vaccination was safe and well tolerated with no major adverse effects. Increased cytotoxic T lymphocyte response and the anti-peptide IgG titre were revealed by the post-vaccination sera in eight patients. • Clinical responses were as follows: one complete response, one partial response, two stable disease and six progressive disease. • Median progression-free survival and overall survival were 3.0 and 8.9 months, respectively. In the four responders, median progression-free survival and overall survival were 21 and 24 months, respectively. CONCLUSIONS • This phase I study showed the safety of and boosted immune responses in response to PPV for advanced UC. • The potential efficacy of 12 consecutive weekly vaccinations with PPV in patients with advanced UC merits further investigation based on these findings.
Collapse
|
29
|
Tan A, De La Peña H, Seifalian AM. The application of exosomes as a nanoscale cancer vaccine. Int J Nanomedicine 2010; 5:889-900. [PMID: 21116329 PMCID: PMC2990382 DOI: 10.2147/ijn.s13402] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Cancer is a leading cause of death globally, and it is predicted and projected to continue rising as life expectancy increases. Although patient survival rates for some forms of cancers are high due to clinical advances in treatment protocols, the search for effective cancer vaccines remains the ultimate Rosetta Stone in oncology. Cervarix®, Gardasil®, and hepatitis B vaccines are currently employed in preventing certain forms of viral cancers. However, they are, strictly speaking, not ‘true’ cancer vaccines as they are prophylactic rather than therapeutic, are only effective against the oncogenic viruses, and do not kill the actual cancer cells. On April 2010, a new prostate cancer vaccine Provenge® (sipuleucel-T) was approved by the US FDA, and it is the first approved therapeutic vaccine that utilizes antigen-presenting cell technology involving dendritic cells in cancer immunotherapy. Recent evidence suggests that the use of nanoscale particles like exosomes in immunotherapy could form a viable basis for the development of novel cancer vaccines, via antigen-presenting cell technology, to prime the immune system to recognize and kill cancer cells. Coupled with nanotechnology, engineered exosomes are emerging as new and novel avenues for cancer vaccine development. Here, we review the current knowledge pertaining to exosome technology in immunotherapy and also seek to address the challenges and future directions associated with it, in hopes of bringing this exciting application a step closer toward an effective clinical reality.
Collapse
Affiliation(s)
- Aaron Tan
- Centre for Nanotechnology and Regenerative Medicine, University College London, London, UK
| | | | | |
Collapse
|