51
|
Hardwick N, Frankel PH, Cristea M. New Approaches for Immune Directed Treatment for Ovarian Cancer. Curr Treat Options Oncol 2016; 17:14. [PMID: 26942589 DOI: 10.1007/s11864-016-0389-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT The immune system plays an active role in the pathogenesis of ovarian cancer (OC), as well as in the mechanisms of disease progression and overall survival (OS). Immunotherapy in gynecological cancers could help to revert immunosuppression and lymphocyte depletion due to prior treatments. Current immunotherapies for ovarian cancer, like all cancer immunotherapy, are based on either stimulating the immune system or reverting immune suppression. Several approaches have been used, including therapeutic vaccines, monoclonal antibodies; checkpoint inhibitors and adoptive T cell transfer. Most of these therapies are still in early-phase testing (phase I and II) for ovarian cancer, but the initial data in ovarian cancer and successful use in other types of cancers suggests some of these approaches may ultimately prove useful for ovarian cancer as well. Ovarian cancer vaccines have shown only a modest benefit in ovarian cancer when used as monotherapy, but these agents may be able to enhance antitumor activity when combined with chemotherapy, checkpoint inhibitors, or other immunotherapies. Monoclonal antibodies have been explored in ovarian cancer but despite encouraging phase II data, randomized studies failed to demonstrate significant clinical benefit. Check point inhibitors have promising activity in several solid tumors and have demonstrated a favorable toxicity profile. Data from early clinical trials utilizing PD1 and PD-L1 inhibitors showed encouraging results. Ongoing clinical trials are evaluating the role of check point inhibitors in combination with chemotherapy. Adoptive T cell transfer involves the infusion of ex vivo activated and expanded tumor specific T cells, using various sources and types of T cells. While this approach has been explored in several hematologic malignancies, it constitutes early research in ovarian cancer. Immunotherapy remains investigational in ovarian cancer and the benefit of this approach in improving progression-free survival (PFS) or OS is unknown. Previous clinical trials have not selected patients based on biomarkers and this may explain the negative results. We expect to discover that tumor response will relate to the patient's immune features and specific tumor characteristics. We are only beginning to realize the potential of immunotherapy for ovarian cancer patients, and one goal of future clinical trials will be to identify subsets of patient based on histologic, molecular, and immune characteristics.
Collapse
Affiliation(s)
- Nicola Hardwick
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Paul H Frankel
- Division of Biostatistics, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Mihaela Cristea
- Department of Medical Oncology, City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
52
|
Yu Z, Tan Z, Lee BK, Tang J, Wu X, Cheung KW, Lo NTL, Man K, Liu L, Chen Z. Antigen spreading-induced CD8+T cells confer protection against the lethal challenge of wild-type malignant mesothelioma by eliminating myeloid-derived suppressor cells. Oncotarget 2016; 6:32426-38. [PMID: 26431275 PMCID: PMC4741703 DOI: 10.18632/oncotarget.5856] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/06/2015] [Indexed: 12/02/2022] Open
Abstract
A key focus in cancer immunotherapy is to investigate the mechanism of efficacious vaccine responses. Using HIV-1 GAG-p24 in a model PD1-based DNA vaccine, we recently reported that vaccine-elicited CD8+ T cells conferred complete prevention and therapeutic cure of AB1-GAG malignant mesothelioma in immunocompetent BALB/c mice. Here, we further investigated the efficacy and correlation of protection on the model vaccine-mediated antigen spreading against wild-type AB1 (WT-AB1) mesothelioma. We found that this vaccine was able to protect mice completely from three consecutive lethal challenges of AB1-GAG mesothelioma. Through antigen spreading these animals also developed tumor-specific cytotoxic CD8+ T cells, but neither CD4+ T cells nor antibodies, rejecting WT-AB1 mesothelioma. A majority of these protected mice (90%) were also completely protected against the lethal WT-AB1 challenge. Adoptive cell transfer experiments further demonstrated that antigen spreading-induced CD8+ T cells conferred efficacious therapeutic effects against established WT-AB1 mesothelioma and prevented the increase of exhausted PD-1+ and Tim-3+ CD8+ T cells. A significant inverse correlation was found between the frequency of functional PD1−Tim3− CD8+ T cells and that of MDSCs or tumor mass in vivo. Mechanistically, we found that WT-AB1 mesothelioma induced predominantly polymorphonuclear (PMN) MDSCs in vivo. In co-cultures with efficacious CD8+ T cells, a significant number of PMN-MDSCs underwent apoptosis in a dose-dependent way. Our findings indicate that efficacious CD8+ T cells capable of eliminating both tumor cells and MDSCs are likely necessary for fighting wild-type malignant mesothelioma.
Collapse
Affiliation(s)
- Zhe Yu
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China.,Department of Orthopedic Surgery, Orthopedics Oncology Institute of Chinese PLA, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, P.R. China
| | - Zhiwu Tan
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Boon Kiat Lee
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Jiansong Tang
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Xilin Wu
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Ka-Wai Cheung
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Nathan Tin Lok Lo
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Kwan Man
- Department of Surgery and Centre for Cancer Research, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Li Liu
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Zhiwei Chen
- AIDS Institute and Department of Microbiology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China.,Research Center for Infection and Immunity, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China
| |
Collapse
|
53
|
Huang Y, Ma C, Zhang Q, Ye J, Wang F, Zhang Y, Hunborg P, Varvares MA, Hoft DF, Hsueh EC, Peng G. CD4+ and CD8+ T cells have opposing roles in breast cancer progression and outcome. Oncotarget 2016; 6:17462-78. [PMID: 25968569 PMCID: PMC4627321 DOI: 10.18632/oncotarget.3958] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/09/2015] [Indexed: 12/13/2022] Open
Abstract
The Cancer Immunoediting concept has provided critical insights suggesting dual functions of immune system during the cancer initiation and development. However, the dynamics and roles of CD4+ and CD8+ T cells in the pathogenesis of breast cancer remain unclear. Here we utilized two murine breast cancer models (4T1 and E0771) and demonstrated that both CD4+ and CD8+ T cells were increased and involved in immune responses, but with distinct dynamic trends in breast cancer development. In addition to cell number increases, CD4+ T cells changed their dominant subsets from Th1 in the early stages to Treg and Th17 cells in the late stages of the cancer progression. We also analyzed CD4+ and CD8+ T cell infiltration in primary breast cancer tissues from cancer patients. We observed that CD8+ T cells are the key effector cell population mediating effective anti-tumor immunity resulting in better clinical outcomes. In contrast, intra-tumoral CD4+ T cells have negative prognostic effects on breast cancer patient outcomes. These studies indicate that CD4+ and CD8+ T cells have opposing roles in breast cancer progression and outcomes, which provides new insights relevant for the development of effective cancer immunotherapeutic approaches.
Collapse
Affiliation(s)
- Yi Huang
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA.,Center for Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Chunling Ma
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA.,Department of Laboratory Medicine, Women and Children's Health Care Hospital of Linyi City, Linyi, P. R. China.,Molecular Biology Experimental Center, Shandong Medical College, Linyi, P. R. China
| | - Qunyuan Zhang
- Department of Genetics, Washington University School of Medicine in St. Louis, Saint Louis, MO, USA
| | - Jian Ye
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Fang Wang
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA.,Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P. R. China
| | - Yanping Zhang
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Pamela Hunborg
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Mark A Varvares
- Department of Otolaryngology-Head and Neck Surgery, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Daniel F Hoft
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Eddy C Hsueh
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Guangyong Peng
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
54
|
Stanton SE, Eary JF, Marzbani EA, Mankoff D, Salazar LG, Higgins D, Childs J, Reichow J, Dang Y, Disis ML. Concurrent SPECT/PET-CT imaging as a method for tracking adoptively transferred T-cells in vivo. J Immunother Cancer 2016; 4:27. [PMID: 27190628 PMCID: PMC4869363 DOI: 10.1186/s40425-016-0131-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/15/2016] [Indexed: 12/17/2022] Open
Abstract
Background The ability of T-cells to traffic to and penetrate tumors impacts the clinical efficacy of T-cell therapy therefore methods to track transferred T-cells in vivo are needed. In this preliminary report, we evaluated the use of concurrent SPECT/PET-CT imaging to monitor the egress of HER-2/neu specific T-cells in a breast cancer patient with extensive bone-only metastatic disease. Findings Indium (In-111) labeled T-cells demonstrated similar or greater viability than unlabeled T-cells at either a low or high dose of In-111 over a 24-h incubation period in vitro. The function of labeled or unlabeled T-cells was not significantly different (p > 0.05) at either dose. T-cells trafficked to all sites of metastatic disease and infiltrated the tumor as assessed by SPECT imaging. In-111 uptake at 24 h after infusion varied from 3.8 (right proximal humerus) to 6.3 (right sacrum) background corrected counts per pixel and remained elevated at 48 h. Concurrent PET-CT imaging demonstrated a fluorodeoxyglucose flare, measured by increase in tumor site uptake as high as 32 % and at most sites of disease at 48 h. This flare was associated with focal pain after T-cell infusion at metastatic sites. The patient had stable disease for 18 months after completion of T-cell therapy. Conclusion Concurrent SPECT/PET-CT imaging, over a 48-h period after T-cell infusion, provided evidence of T-cell homing to all disease sites as well as a tumor metabolism flare response. This technique may be useful for monitoring T-cell trafficking after autologous as well as chimeric antigen receptor T-cell infusion. Trial Registraion Trial registered at ClinicalTrials.gov registration number NCT00791037, registered 13 November 2008.
Collapse
Affiliation(s)
- Sasha E Stanton
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Janet F Eary
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35249 USA
| | - Edmond A Marzbani
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - David Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Lupe G Salazar
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Doreen Higgins
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Jennifer Childs
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Jessica Reichow
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Yushe Dang
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109 USA
| |
Collapse
|
55
|
Martin Lluesma S, Wolfer A, Harari A, Kandalaft LE. Cancer Vaccines in Ovarian Cancer: How Can We Improve? Biomedicines 2016; 4:biomedicines4020010. [PMID: 28536377 PMCID: PMC5344251 DOI: 10.3390/biomedicines4020010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 04/15/2016] [Accepted: 04/19/2016] [Indexed: 12/11/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is one important cause of gynecologic cancer-related death. Currently, the mainstay of ovarian cancer treatment consists of cytoreductive surgery and platinum-based chemotherapy (introduced 30 years ago) but, as the disease is usually diagnosed at an advanced stage, its prognosis remains very poor. Clearly, there is a critical need for new treatment options, and immunotherapy is one attractive alternative. Prophylactic vaccines for prevention of infectious diseases have led to major achievements, yet therapeutic cancer vaccines have shown consistently low efficacy in the past. However, as they are associated with minimal side effects or invasive procedures, efforts directed to improve their efficacy are being deployed, with Dendritic Cell (DC) vaccination strategies standing as one of the more promising options. On the other hand, recent advances in our understanding of immunological mechanisms have led to the development of successful strategies for the treatment of different cancers, such as immune checkpoint blockade strategies. Combining these strategies with DC vaccination approaches and introducing novel combinatorial designs must also be considered and evaluated. In this review, we will analyze past vaccination methods used in ovarian cancer, and we will provide different suggestions aiming to improve their efficacy in future trials.
Collapse
Affiliation(s)
- Silvia Martin Lluesma
- Center of Experimental Therapeutics, Ludwig Center for Cancer Res, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Anita Wolfer
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Alexandre Harari
- Center of Experimental Therapeutics, Ludwig Center for Cancer Res, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Ludwig Center for Cancer Res, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
56
|
Knutson KL, Clynes R, Shreeder B, Yeramian P, Kemp KP, Ballman K, Tenner KS, Erskine CL, Norton N, Northfelt D, Tan W, Calfa C, Pegram M, Mittendorf EA, Perez EA. Improved Survival of HER2+ Breast Cancer Patients Treated with Trastuzumab and Chemotherapy Is Associated with Host Antibody Immunity against the HER2 Intracellular Domain. Cancer Res 2016; 76:3702-10. [PMID: 27197192 DOI: 10.1158/0008-5472.can-15-3091] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/06/2016] [Indexed: 01/16/2023]
Abstract
The addition of trastuzumab to chemotherapy extends survival among patients with HER2(+) breast cancer. Prior work showed that trastuzumab and chemotherapy augments HER2 extracellular domain (ECD)-specific antibodies. The current study investigated whether combination therapy induced immune responses beyond HER2-ECD and, importantly, whether those immune responses were associated with survival. Pretreatment and posttreatment sera were obtained from 48 women with metastatic HER2(+) breast cancer on NCCTG (now Alliance for Clinical Trials in Oncology) studies, N0337 and N983252. IgG to HER2 intracellular domain (ICD), HER2-ECD, p53, IGFBP2, CEA, and tetanus toxoid were examined. Sera from 25 age-matched controls and 26 surgically resected HER2(+) patients were also examined. Prior to therapy, some patients with metastatic disease had elevated antibodies to IGFBP2, p53, HER2-ICD, HER2-ECD, and CEA, but not to tetanus toxin, relative to controls and surgically resected patients. Treatment augmented antibody responses to HER2-ICD in 69% of metastatic patients, which was highly associated with improved progression-free survival (PFS; HR = 0.5, P = 0.0042) and overall survival (OS; HR = 0.7, P = 0.038). Augmented antibody responses to HER2-ICD also correlated (P = 0.03) with increased antibody responses to CEA, IGFBP2, and p53, indicating that treatment induces epitope spreading. Paradoxically, patients who already had high preexisting immunity to HER2-ICD did not respond to therapy with increased antibodies to HER2-ICD and demonstrated poorer PFS (HR = 1.6, P < 0.0001) and OS (HR = 1.4, P = 0.0006). Overall, the findings further demonstrate the importance of the adaptive immune system in the efficacy of trastuzumab-containing regimens. Cancer Res; 76(13); 3702-10. ©2016 AACR.
Collapse
Affiliation(s)
- Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, Florida.
| | - Raphael Clynes
- Division of Hematology and Oncology, Columbia University, New York, New York
| | - Barath Shreeder
- Department of Immunology, Mayo Clinic, Jacksonville, Florida
| | - Patrick Yeramian
- Cancer Vaccines and Immune Therapies Program, Center for Diseases of Aging, Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida
| | - Kathleen P Kemp
- Cancer Vaccines and Immune Therapies Program, Center for Diseases of Aging, Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida
| | - Karla Ballman
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Kathleen S Tenner
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Nadine Norton
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Donald Northfelt
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Winston Tan
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida
| | | | | | - Elizabeth A Mittendorf
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edith A Perez
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
57
|
Clifton GT, Peoples GE, Mittendorf EA. The development and use of the E75 (HER2 369-377) peptide vaccine. Future Oncol 2016; 12:1321-9. [PMID: 27044454 DOI: 10.2217/fon-2015-0054] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
E75 (nelipepimut-S) is an immunogenic peptide derived from the HER2 protein. When combined with the immunoadjuvant granulocyte-macrophage colony-stimulating factor (GM-CSF), nelipepimut-S has been used as a vaccine that is capable of eliciting a robust anti-HER2 immune response. Early-phase clinical trials that enrolled women with node-positive or high-risk node-negative breast cancer who had been rendered disease free with standard of care therapy but were at risk for recurrence, demonstrated the vaccine to be safe with a suggestion of clinical benefit. Nelipepimut-S is currently being evaluated in a Phase III clinical trial. This article covers the preclinical and clinical development of nelipepimut-S.
Collapse
Affiliation(s)
- Guy T Clifton
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA
| | - George E Peoples
- Cancer Vaccine Development Program, Metis Foundation, 600 Navarro Street, San Antonio, TX 78205, USA
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1434, Houston, TX 77030, USA
| |
Collapse
|
58
|
Yung MMH, Ngan HYS, Chan DW. Targeting AMPK signaling in combating ovarian cancers: opportunities and challenges. Acta Biochim Biophys Sin (Shanghai) 2016; 48:301-17. [PMID: 26764240 PMCID: PMC4886241 DOI: 10.1093/abbs/gmv128] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/29/2015] [Indexed: 12/25/2022] Open
Abstract
The development and strategic application of effective anticancer therapies have turned out to be one of the most critical approaches of managing human cancers. Nevertheless, drug resistance is the major obstacle for clinical management of these diseases especially ovarian cancer. In the past years, substantial studies have been carried out with the aim of exploring alternative therapeutic approaches to enhance efficacy of current chemotherapeutic regimes and reduce the side effects caused in order to produce significant advantages in overall survival and to improve patients' quality of life. Targeting cancer cell metabolism by the application of AMP-activated protein kinase (AMPK)-activating agents is believed to be one of the most plausible attempts. AMPK activators such as 5-aminoimidazole-4-carboxamide 1-β-d-ribofuranoside, A23187, metformin, and bitter melon extract not only prevent cancer progression and metastasis but can also be applied as a supplement to enhance the efficacy of cisplatin-based chemotherapy in human cancers such as ovarian cancer. However, because of the undesirable outcomes along with the frequent toxic side effects of most pharmaceutical AMPK activators that have been utilized in clinical trials, attentions of current studies have been aimed at the identification of replaceable reagents from nutraceuticals or traditional medicines. However, the underlying molecular mechanisms of many nutraceuticals in anticancer still remain obscure. Therefore, better understanding of the functional characterization and regulatory mechanism of natural AMPK activators would help pharmaceutical development in opening an area to intervene ovarian cancer and other human cancers.
Collapse
Affiliation(s)
- Mingo M H Yung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hextan Y S Ngan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - David W Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
59
|
Clifton GT, Gall V, Peoples GE, Mittendorf EA. Clinical Development of the E75 Vaccine in Breast Cancer. Breast Care (Basel) 2016; 11:116-21. [PMID: 27239173 PMCID: PMC4881244 DOI: 10.1159/000446097] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
E75 is an immunogenic peptide derived from the human epidermal growth factor receptor 2 (HER2) protein. A large amount of preclinical work evaluated the immunogenicity of E75, after which phase I trials investigated using E75 mixed with an immunoadjuvant as a vaccine. Those studies showed the vaccine to be safe and capable of stimulating an antigen-specific immune response. Subsequent to that, our group conducted trials evaluating E75 + granulocyte macrophage colony-stimulating factor (GM-CSF) in the adjuvant setting. The studies enrolled node-positive and high-risk node-negative breast cancer patients, with the goal being to determine if vaccination could decrease the recurrence risk. The studies included 187 evaluable patients: 108 vaccinated ones and 79 controls. The 5-year disease-free survival for the vaccinated patients was 89.7% compared to 80.2% for the control patients, a 48% reduction in relative risk of recurrence. Based on these data, E75 + GM-CSF, now known as NeuVax™, is being evaluated in a phase III trial. In this article, we review preclinical data and results of the early-phase trials and provide an update on the ongoing phase III study. We also present additional strategies for employing the vaccine to be included as a component of combination immunotherapy as well as in the setting of ductal carcinoma in situ as an initial step towards primary prevention.
Collapse
Affiliation(s)
- Guy T. Clifton
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victor Gall
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George E. Peoples
- Cancer Vaccine Development Program, Metis Foundation, San Antonio, TX, USA
| | - Elizabeth A. Mittendorf
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
60
|
A non-randomized dose-escalation Phase I trial of a protein-based immunotherapeutic for the treatment of breast cancer patients with HER2-overexpressing tumors. Breast Cancer Res Treat 2016; 156:319-30. [DOI: 10.1007/s10549-016-3751-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/07/2016] [Indexed: 11/26/2022]
|
61
|
Antonilli M, Rahimi H, Visconti V, Napoletano C, Ruscito I, Zizzari IG, Caponnetto S, Barchiesi G, Iadarola R, Pierelli L, Rughetti A, Bellati F, Panici PB, Nuti M. Triple peptide vaccination as consolidation treatment in women affected by ovarian and breast cancer: Clinical and immunological data of a phase I/II clinical trial. Int J Oncol 2016; 48:1369-78. [PMID: 26892612 PMCID: PMC4777599 DOI: 10.3892/ijo.2016.3386] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/13/2016] [Indexed: 12/21/2022] Open
Abstract
Vaccination with priming and expansion of tumour reacting T cells is an important therapeutic option to be used in combination with novel checkpoint inhibitors to increase the specificity of the T cell infiltrate and the efficacy of the treatment. In this phase I/II study, 14 high-risk disease-free ovarian (OC) and breast cancer (BC) patients after completion of standard therapies were vaccinated with MUC1, ErbB2 and carcinoembryonic antigen (CEA) HLA-A2+-restricted peptides and Montanide. Patients were subjected to 6 doses of vaccine every two weeks and a recall dose after 3 months. ECOG grade 2 toxicity was observed at the injection site. Eight out of 14 patients showed specific CD8+ T cells to at least one antigen. None of 4 patients vaccinated for compassionate use showed a CD8 activation. An OC patient who suffered from a lymph nodal recurrence, showed specific anti-ErbB2 CD8+ T cells in the bulky aortic lymph nodes suggesting homing of the activated T cells. Results confirm that peptide vaccination strategy is feasible, safe and well tolerated. In particular OC patients appear to show a higher response rate compared to BC patients. Vaccination generates a long-lasting immune response, which is strongly enhanced by recall administrations. The clinical outcome of patients enrolled in the trial appears favourable, having registered no deceased patients with a minimum follow-up of 8 years. These promising data, in line with the results of similar studies, the high compliance of patients observed and the favourable toxicity profile, support future trials of peptide vaccination in clinically disease-free patients who have completed standard treatments.
Collapse
Affiliation(s)
- Morena Antonilli
- Department of Gynecology, Obstetrics and Urology, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Hassan Rahimi
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Valeria Visconti
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Chiara Napoletano
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Ilary Ruscito
- Department of Gynecology, Obstetrics and Urology, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Ilaria Grazia Zizzari
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Salvatore Caponnetto
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Giacomo Barchiesi
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Roberta Iadarola
- Department of Gynecology, Obstetrics and Urology, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Luca Pierelli
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Aurelia Rughetti
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Filippo Bellati
- Department of Gynecology, Obstetrics and Urology, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Pierluigi Benedetti Panici
- Department of Gynecology, Obstetrics and Urology, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine, 'Sapienza' University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| |
Collapse
|
62
|
Drerup JM, Liu Y, Padron AS, Murthy K, Hurez V, Zhang B, Curiel TJ. Immunotherapy for ovarian cancer. Curr Treat Options Oncol 2015; 16:317. [PMID: 25648541 DOI: 10.1007/s11864-014-0317-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OPINION STATEMENT All work referenced herein relates to treatment of epithelial ovarian carcinomas, as their treatment differs from ovarian germ cell cancers and other rare ovarian cancers, the treatments of which are addressed elsewhere. Fallopian tube cancers and primary peritoneal adenocarcinomatosis are also generally treated as epithelial ovarian cancers. The standard of care initial treatment of advanced stage epithelial ovarian cancer is optimal debulking surgery as feasible plus chemotherapy with a platinum plus a taxane agent. If this front-line approach fails, as it too often the case, several FDA-approved agents are available for salvage therapy. However, because no second-line therapy for advanced-stage epithelial ovarian cancer is typically curative, we prefer referral to clinical trials as logistically feasible, even if it means referring patients outside our system. Immune therapy has a sound theoretical basis for treating carcinomas generally, and for treating ovarian cancer in particular. Advances in understanding the immunopathogenic basis of ovarian cancer, and the immunopathologic basis for prior failures of immunotherapy for it and other carcinomas promises to afford novel treatment approaches with potential for significant efficacy, and reduced toxicities compared with cytotoxic agents. Thus, referral to early phase immunotherapy trials for ovarian cancer patients that fail conventional treatment merits consideration.
Collapse
Affiliation(s)
- Justin M Drerup
- Department of Cellular and Structural Biology, School of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | | | | | | | | | | | | |
Collapse
|
63
|
Peper JK, Bösmüller HC, Schuster H, Gückel B, Hörzer H, Roehle K, Schäfer R, Wagner P, Rammensee HG, Stevanović S, Fend F, Staebler A. HLA ligandomics identifies histone deacetylase 1 as target for ovarian cancer immunotherapy. Oncoimmunology 2015; 5:e1065369. [PMID: 27467910 PMCID: PMC4910750 DOI: 10.1080/2162402x.2015.1065369] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 10/28/2022] Open
Abstract
The recent approval of clincially effective immune checkpoint inhibitors illustrates the potential of cancer immunotherapy. A challenging task remains the identification of specific targets guiding immunotherapy. Facilitated by technical advances, the direct identification of physiologically relevant targets is enabled by analyzing the HLA ligandome of cancer cells. Since recent publications demonstrate the immunogenicity of ovarian cancer (OvCa), immunotherapies, including peptide-based cancer vaccines, represent a promising treatment approach. To identify vaccine peptides, we employed a combined strategy of HLA ligandomics in high-grade serous OvCa samples and immunogenicity analysis. Only few proteins were naturally presented as HLA ligands on all samples analyzed, including histone deacetylase (HDAC) 1 and 2. In vitro priming of CD8(+) T cells demonstrated that two HDAC1/2-derived HLA ligands can induce T-cell responses, capable of killing HLA-matched tumor cells. High HDAC1 expression shown by immunohistochemistry in 136 high-grade serous OvCa patients associated with significantly reduced overall survival (OS), whereas patients with high numbers of CD3(+) tumor-infiltrating lymphocytes (TILs) in the tumor epithelium and CD8(+) TILs in the tumor stroma showed improved OS. However, correlating HDAC1 expression with TILs, high levels of TILs abrogated the impact of HDAC1 on OS. This study strengthens the role of HDAC1/2 as an important tumor antigen in OvCa, demonstrating its impact on OS in a large cohort of OvCa patients. We further identified two immunogenic HDAC1-derived peptides, which frequently induce multi-functional T-cell responses in many donors, suitable for future multi-peptide vaccine trials in OvCa patients.
Collapse
Affiliation(s)
- Janet Kerstin Peper
- Department of Immunology, Institute of Cell Biology, University of Tübingen , Tübingen, Germany
| | | | - Heiko Schuster
- Department of Immunology, Institute of Cell Biology, University of Tübingen , Tübingen, Germany
| | - Brigitte Gückel
- Department of Obstetrics and Gynecology, University Hospital of Tübingen , Tübingen, Germany
| | - Helen Hörzer
- Department of Obstetrics and Gynecology, University Hospital of Tübingen , Tübingen, Germany
| | - Kevin Roehle
- Department of Immunology, Institute of Cell Biology, University of Tübingen , Tübingen, Germany
| | - Richard Schäfer
- Department of Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen , Tübingen, Germany
| | - Philipp Wagner
- Department of Obstetrics and Gynecology, University Hospital of Tübingen , Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute of Cell Biology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- Department of Immunology, Institute of Cell Biology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Falko Fend
- Institute of Pathology, University Hospital of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Annette Staebler
- Institute of Pathology, University Hospital of Tübingen , Tübingen, Germany
| |
Collapse
|
64
|
Galaine J, Borg C, Godet Y, Adotévi O. Interest of Tumor-Specific CD4 T Helper 1 Cells for Therapeutic Anticancer Vaccine. Vaccines (Basel) 2015; 3:490-502. [PMID: 26350591 PMCID: PMC4586463 DOI: 10.3390/vaccines3030490] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 12/16/2022] Open
Abstract
Nowadays, immunotherapy represents one promising approach for cancer treatment. Recently, spectacular results of cancer immunotherapy clinical trials have confirmed the crucial role of immune system in cancer regression. Therapeutic cancer vaccine represents one widely used immunotherapy strategy to stimulate tumor specific T cell responses but clinical impact remains disappointing in targeting CD8 T cells. Although CD8 T cells have been initially considered to be the main protagonists, it is now clear that CD4 T cells also play a critical role in antitumor response. In this article, we discuss the role of tumor antigen-specific CD4 T cell responses and how we can target these cells to improve cancer vaccines.
Collapse
Affiliation(s)
- Jeanne Galaine
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
| | - Christophe Borg
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
- Department of Medical Oncology, University Hospital of Besançon, Besançon cedex F25020, France.
| | - Yann Godet
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
| | - Olivier Adotévi
- INSERM UMR1098, Besançon cedex F25020, France.
- Université de Franche-Comté, Besançon cedex F25020, France.
- EFS Bourgogne Franche-Comté, Besançon cedex F25020, France.
- Department of Medical Oncology, University Hospital of Besançon, Besançon cedex F25020, France.
| |
Collapse
|
65
|
Abstract
The toxicities of immunotherapy for cancer are as diverse as the type of treatments that have been devised. These range from cytokine therapies that induce capillary leakage to vaccines associated with low levels of autoimmunity to cell therapies that can induce damaging cross-reactivity with normal tissue to checkpoint protein inhibitors that induce immune-related adverse events that are autoinflammatory in nature. The thread that ties these toxicities together is their mechanism-based immune nature and the T-cell-mediated adverse events seen. The basis for the majority of these adverse events is a hyperactivated T-cell response with reactivity directed against normal tissue, resulting in the generation of high levels of CD4 T-helper cell cytokines or increased migration of cytolytic CD8 T cells within normal tissues. The T-cell immune response is not tissue specific and may reflect a diffuse expansion of the T-cell repertoire that induces cross-reactivity with normal tissue, effectively breaking tolerance that is active with cytokines, vaccines, and checkpoint protein inhibitors and passive in the case of adoptive cell therapy. Cytokines seem to generate diffuse and nonspecific T-cell reactivity, whereas checkpoint protein inhibition, vaccines, and adoptive cell therapy seem to activate more specific T cells that interact directly with normal tissues, potentially causing specific organ damage. In this review, we summarize the toxicities that are unique to immunotherapies, emphasizing the need to familiarize the oncology practitioner with the spectrum of adverse events seen with newly approved and emerging modalities.
Collapse
Affiliation(s)
- Jeffrey S Weber
- Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; James C. Yang, National Cancer Institute, Bethesda, MD; Michael B. Atkins, Lombardi Cancer Center, Georgetown University, Washington, DC; and Mary L. Disis, The Fred Hutchinson Cancer Center, University of Washington, Seattle, WA.
| | - James C Yang
- Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; James C. Yang, National Cancer Institute, Bethesda, MD; Michael B. Atkins, Lombardi Cancer Center, Georgetown University, Washington, DC; and Mary L. Disis, The Fred Hutchinson Cancer Center, University of Washington, Seattle, WA
| | - Michael B Atkins
- Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; James C. Yang, National Cancer Institute, Bethesda, MD; Michael B. Atkins, Lombardi Cancer Center, Georgetown University, Washington, DC; and Mary L. Disis, The Fred Hutchinson Cancer Center, University of Washington, Seattle, WA
| | - Mary L Disis
- Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; James C. Yang, National Cancer Institute, Bethesda, MD; Michael B. Atkins, Lombardi Cancer Center, Georgetown University, Washington, DC; and Mary L. Disis, The Fred Hutchinson Cancer Center, University of Washington, Seattle, WA
| |
Collapse
|
66
|
Patel JM, Vartabedian VF, Kim MC, He S, Kang SM, Selvaraj P. Influenza virus-like particles engineered by protein transfer with tumor-associated antigens induces protective antitumor immunity. Biotechnol Bioeng 2015; 112:1102-10. [PMID: 25689082 PMCID: PMC4621003 DOI: 10.1002/bit.25537] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/18/2014] [Accepted: 12/30/2014] [Indexed: 01/06/2023]
Abstract
Delivery of antigen in particulate form using either synthetic or natural particles induces stronger immunity than soluble forms of the antigen. Among naturally occurring particles, virus-like particles (VLPs) have been genetically engineered to express tumor-associated antigens (TAAs) and have shown to induce strong TAA-specific immune responses due to their nano-particulate size and ability to bind and activate antigen-presenting cells. In this report, we demonstrate that influenza VLPs can be modified by a protein transfer technology to express TAAs for induction of effective antitumor immune responses. We converted the breast cancer HER-2 antigen to a glycosylphosphatidylinositol (GPI)-anchored form and incorporated GPI-HER-2 onto VLPs by a rapid protein transfer process. Expression levels on VLPs depended on the GPI-HER-2 concentration added during protein transfer. Vaccination of mice with protein transferred GPI-HER-2-VLPs induced a strong Th1 and Th2-type anti-HER-2 antibody response and protected mice against a HER-2-expressing tumor challenge. The Soluble form of GPI-HER-2 induced only a weak Th2 response under similar conditions. These results suggest that influenza VLPs can be enriched with TAAs by protein transfer to develop effective VLP-based subunit vaccines against cancer without chemical or genetic modifications and thus preserve the immune stimulating properties of VLPs for easier production of antigen-specific therapeutic cancer vaccines.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/blood
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Disease Models, Animal
- Drug Carriers
- Humans
- Immunity
- Mice
- Neoplasms/immunology
- Neoplasms/prevention & control
- Orthomyxoviridae/genetics
- Orthomyxoviridae/metabolism
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Jaina M. Patel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322. USA
| | - Vincent F. Vartabedian
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322. USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
- Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Sara He
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322. USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322. USA
| |
Collapse
|
67
|
Abstract
Cancer vaccines are designed to promote tumor specific immune responses, particularly cytotoxic CD8 positive T cells that are specific to tumor antigens. The earliest vaccines, which were developed in 1994-95, tested non-mutated, shared tumor associated antigens that had been shown to be immunogenic and capable of inducing clinical responses in a minority of people with late stage cancer. Technological developments in the past few years have enabled the investigation of vaccines that target mutated antigens that are patient specific. Several platforms for cancer vaccination are being tested, including peptides, proteins, antigen presenting cells, tumor cells, and viral vectors. Standard of care treatments, such as surgery and ablation, chemotherapy, and radiotherapy, can also induce antitumor immunity, thereby having cancer vaccine effects. The monitoring of patients' immune responses at baseline and after standard of care treatment is shedding light on immune biomarkers. Combination therapies are being tested in clinical trials and are likely to be the best approach to improving patient outcomes.
Collapse
Affiliation(s)
- Lisa H Butterfield
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
68
|
Kazemi T, Younesi V, Jadidi-Niaragh F, Yousefi M. Immunotherapeutic approaches for cancer therapy: An updated review. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:769-79. [PMID: 25801036 DOI: 10.3109/21691401.2015.1019669] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In spite of specific immune effector mechanisms raised against tumor cells, there are mechanisms employed by the tumor cells to keep them away from immune recognition and elimination; some of these mechanisms have been identified, while others are still poorly understood. Manipulation or augmentation of specific antitumor immune responses are now the preferred approaches for treatment of malignancies, and traditional therapeutic approaches are being replaced by the use of agents which potentiate immune effector mechanisms, broadly called "immunotherapy". Cancer immunotherapy is generally classified into two main classes including active and passive methods. Interventions to augment the immune system of the patient, for example, vaccination or adjuvant therapy, actively promote antitumor effector mechanisms to improve cancer elimination. On the other hand, administration of specific monoclonal antibodies (mAbs) against different tumor antigens and adoptive transfer of genetically-modified specific T cells are currently the most rapidly developing approaches for cancer targeted therapy. In this review, we will discuss the different modalities for active and passive immunotherapy for cancer.
Collapse
Affiliation(s)
- Tohid Kazemi
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Vahid Younesi
- c Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Farhad Jadidi-Niaragh
- c Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Yousefi
- a Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
69
|
Schneble EJ, Berry JS, Trappey FA, Clifton GT, Ponniah S, Mittendorf E, Peoples GE. The HER2 peptide nelipepimut-S (E75) vaccine (NeuVax™) in breast cancer patients at risk for recurrence: correlation of immunologic data with clinical response. Immunotherapy 2015; 6:519-31. [PMID: 24896623 DOI: 10.2217/imt.14.22] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Nelipepimut-S (formerly known as E75) is an immunogenic peptide from the HER2 protein that is highly expressed in breast cancer. The NeuVax™ (Galena, OR, USA) vaccine, nelipepimut-S plus granulocyte-macrophage colony-stimulating factor, is designed for the prevention of clinical recurrences in high risk, disease-free breast cancer patients. Although cancer vaccines such as NeuVax represent promising approaches to cancer immunotherapy, much remains to be elucidated regarding their mechanisms of action: particularly given that multiple cancer vaccine trials have failed to demonstrate a correlation between immunologic data and clinical outcome. Here, we briefly discuss our clinical trial experience with NeuVax focusing on immunologic response data and its implication on how the immune system may be affected by this peptide vaccine. Most importantly, we demonstrate the potential capability of certain immunologic assays to predict clinical benefit in our trials.
Collapse
Affiliation(s)
- Erika J Schneble
- San Antonio Military Medical Center, Department of General Surgery, Fort Sam Houston, TX, USA
| | | | | | | | | | | | | |
Collapse
|
70
|
Omabe M, Ahmed S, Sami A, Xie Y, Tao M, Xiang J. HER2-Specific Vaccines for HER2-Positive Breast Cancer Immunotherapy. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/wjv.2015.52013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
71
|
Peptide-Based Vaccination and Induction of CD8+ T-Cell Responses Against Tumor Antigens in Breast Cancer. BioDrugs 2014; 29:15-30. [DOI: 10.1007/s40259-014-0114-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
72
|
Yang J, Zhang Q, Li K, Yin H, Zheng JN. Composite peptide-based vaccines for cancer immunotherapy (Review). Int J Mol Med 2014; 35:17-23. [PMID: 25395173 DOI: 10.3892/ijmm.2014.2000] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 11/03/2014] [Indexed: 11/06/2022] Open
Abstract
The use of peptide-based vaccines as therapeutics aims to elicit immune responses through antigenic epitopes derived from tumor antigens. Peptide-based vaccines are easily synthesized and chemically stable entities, and of note, they are absent of oncogenic potential. However, their application is more complicated as the success of an effective peptide-based vaccine is determined by numerous parameters. The success thus far has been limited by the choice of tumor antigenic peptides, poor immunogenicity and incorporation of strategies to reverse cancer-mediated immune suppression. In the present review, an overview of the mechanisms of peptide-based vaccines is provided and antigenic peptides are categorized with respect to their tissue distribution in order to determine their usefulness as targets. Furthermore, certain approaches are proposed that induce and maintain T cells for immunotherapy. The recent progress indicates that peptide-based vaccines are preferential for targeted therapy in cancer patients.
Collapse
Affiliation(s)
- Jie Yang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| | - Qing Zhang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| | - Ke Li
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| | - Hong Yin
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| | - Jun-Nian Zheng
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
73
|
Baxevanis CN, Anastasopoulou EA, Voutsas IF, Papamichail M, Perez SA. Immune biomarkers: how well do they serve prognosis in human cancers? Expert Rev Mol Diagn 2014; 15:49-59. [PMID: 25345403 DOI: 10.1586/14737159.2015.965684] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In order to be optimally efficacious, therapeutic cancer vaccines must induce robust tumor-specific CD8(+) cytotoxic T cells, which are responsible for tumor cell lysis. Unlike cytotoxic drugs, which act directly on the tumor, cancer vaccines demonstrate new kinetics involving the generation of specific cellular immune responses, which need to be translated into antitumor responses to delay tumor progression and improve survival. These delayed kinetics of action establish a new concept of benefit in the long term, which implies a slow down in tumor growth rates, than a marked reduction in tumor size. Therefore, there is a significant need to identify intermediate biomarkers so that clinical responses can be evaluated in a timely manner. Therapeutic vaccination as a modality for cancer treatment has received significant attention with multiple clinical trials demonstrating improvements in overall survival. Significant challenges to this modality remain, including increasing vaccine potency and minimizing treatment-related toxicities and identifying prognostic and predictive biomarkers of clinical benefit that may guide to select and optimize the therapeutic strategies for patients most likely to gain benefit.
Collapse
Affiliation(s)
- Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras avenue, Athens 11522, Greece
| | | | | | | | | |
Collapse
|
74
|
Milani A, Sangiolo D, Aglietta M, Valabrega G. Recent advances in the development of breast cancer vaccines. BREAST CANCER-TARGETS AND THERAPY 2014; 6:159-68. [PMID: 25339848 PMCID: PMC4204811 DOI: 10.2147/bctt.s38428] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The manipulation of the immune system through the administration of a vaccine to direct an effective and long-lasting immune response against breast cancer (BC) cells is an attractive strategy. Vaccines would have several theoretical advantages over standard therapies, including low toxicities, high specificity, and long-lasting efficacy due to the establishment of immunological memory. However, BC vaccines have failed to demonstrate meaningful results in clinical trials so far. This reflects the intrinsic difficulty in breaking the complex immune-escaping mechanisms developed by cancer cells. New vaccines should be able to elicit complex immunologic response involving multiple immune effectors such as cytotoxic and antibody-secreting B cells, innate immunity effectors, and memory cells. Moreover, especially in patients with large tumor burdens and metastatic disease, combining vaccines with other strategies, such as systemic BC therapies, passive immunotherapy, or immunomodulatory agents, could increase the effectiveness of each approach. Here, we review recent advances in BC vaccines, focusing on suitable targets and innovative strategies. We report results of most recent trials investigating active immunotherapy in BC and provide possible future perspectives in this field of research.
Collapse
Affiliation(s)
- Andrea Milani
- Department of Oncology, University of Torino, Torino, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Torino, Italy
| | - Massimo Aglietta
- Department of Oncology, University of Torino, Torino, Italy ; FPO, Candiolo Cancer Institute, IRCCS, Torino, Italy
| | - Giorgio Valabrega
- Department of Oncology, University of Torino, Torino, Italy ; FPO, Candiolo Cancer Institute, IRCCS, Torino, Italy
| |
Collapse
|
75
|
Subbiah IM, Gonzalez-Angulo AM. Advances and future directions in the targeting of HER2-positive breast cancer: implications for the future. Curr Treat Options Oncol 2014; 15:41-54. [PMID: 24323591 DOI: 10.1007/s11864-013-0262-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OPINION STATEMENT The natural history of HER2-positive breast cancer significantly changed in the past 15 years. Form being the most aggressive type of breast cancer, it became treatable with important cure rates. However, with new and successful drugs, resistance emerges. Progress in research and drug development continues to make available effective anti-HER2 therapies. Our challenge today is to use these tools correctly by looking at the data that support the indications of each compound and to continue clinical trial participation.
Collapse
Affiliation(s)
- Ishwaria M Subbiah
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030-4009, USA
| | | |
Collapse
|
76
|
Leffers N, Daemen T, Helfrich W, Boezen HM, Cohlen BJ, Melief CJM, Nijman HW. Antigen-specific active immunotherapy for ovarian cancer. Cochrane Database Syst Rev 2014:CD007287. [PMID: 25229990 DOI: 10.1002/14651858.cd007287.pub3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Despite advances in chemotherapy, prognosis of ovarian cancer remains poor. Antigen-specific active immunotherapy aims to induce tumour-antigen-specific anti-tumour immune responses as an alternative treatment for ovarian cancer. OBJECTIVES To assess the feasibility of antigen-specific active immunotherapy for ovarian cancer. Primary outcomes are clinical efficacy and antigen-specific immunogenicity with carrier-specific immunogenicity and side effects as secondary outcomes. SEARCH METHODS For the previous version of this review, a systematic search of the Cochrane Central Register of Controlled Trials (CENTRAL) 2009, Issue 3, Cochrane Gynaecological Cancer Group Specialized Register, MEDLINE and EMBASE databases and clinicaltrials.gov was performed (1966 to July 2009). We conducted handsearches of the proceedings of relevant annual meetings (1996 to July 2009).For this update of the review the searches were extended to October 2013. SELECTION CRITERIA Randomised controlled trials (RCTs), as well as non-randomised non-controlled studies that included participants with epithelial ovarian cancer, irrespective of stage of disease, and treated with antigen-specific active immunotherapy, irrespective of type of vaccine, antigen used, adjuvant used, route of vaccination, schedule, and reported clinical or immunological outcomes. DATA COLLECTION AND ANALYSIS Two reviews authors independently performed the data extraction. Risk of bias was evaluated for RCTs according to standard methodological procedures expected by The Cochrane Collabororation or for non-RCTs using a selection of quality domains deemed best applicable to the non-randomised non-controlled studies. MAIN RESULTS Fifty-five studies were included (representing 3051 women with epithelial ovarian cancer). Response definitions showed substantial variation between trials, which makes comparison of trial results unreliable. Information on adverse events was frequently limited. Furthermore, reports of both RCTs and non-RCTs frequently lacked the relevant information necessary to assess risk of bias. Serious biases in most of the included trials can therefore not be ruled out.The largest body of evidence is currently available for CA-125 targeted antibody therapy (16 studies: 2339 participants). Non-RCTs of CA-125 targeted antibody therapy suggests increased survival in humoral and/or cellular responders. However, four large randomised placebo-controlled trials did not show any clinical benefit despite induction of immune responses in approximately 60% of participants.Other small studies targeting many different tumour antigens showed promising immunological results. As these strategies have not yet been tested in RCTs, no reliable inferences about clinical efficacy can be made. Given the promising immunological results, limited side effects and toxicity exploration of clinical efficacy in large well-designed RCTs may be worthwhile. AUTHORS' CONCLUSIONS We conclude that despite promising immunological responses, no clinically effective antigen-specific active immunotherapy is yet available for ovarian cancer. Results should be interpreted cautiously as there was a significant lack of relevant information for the assessment of risk of bias in both RCTs and non-RCTs.
Collapse
Affiliation(s)
- Ninke Leffers
- Obstetrics & Gynecology CB30, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30.001, Groningen, Netherlands, 9700 RB
| | | | | | | | | | | | | |
Collapse
|
77
|
Mittendorf EA, Clifton GT, Holmes JP, Schneble E, van Echo D, Ponniah S, Peoples GE. Final report of the phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer patients. Ann Oncol 2014; 25:1735-1742. [PMID: 24907636 PMCID: PMC4143091 DOI: 10.1093/annonc/mdu211] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 05/24/2014] [Accepted: 05/27/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND E75 (nelipepimut-S) is a human leukocyte antigen (HLA)-A2/A3-restricted immunogenic peptide derived from the HER2 protein. We have conducted phase I/II clinical trials vaccinating breast cancer patients with nelipepimut-S and granulocyte-macrophage colony-stimulating factor (GM-CSF) in the adjuvant setting to prevent disease recurrence. All patients have completed 60 months follow-up, and here, we report the final analyses. PATIENTS AND METHODS The studies were conducted as dose escalation/schedule optimization trials enrolling node-positive and high-risk node-negative patients with tumors expressing any degree of HER2 (immunohistochemistry 1-3+). HLA-A2/3+ patients were vaccinated; others were followed prospectively as controls. Local and systemic toxicity was monitored. Clinical recurrences were documented, and disease-free survival (DFS) was analyzed by Kaplan-Meier curves; groups were compared using log-rank tests. RESULTS Of 195 enrolled patients, 187 were assessable: 108 (57.8%) in the vaccinated group (VG) and 79 (42.2%) in the control group (CG). The groups were well matched for clinicopathologic characteristics. Toxicities were minimal. Five-year DFS was 89.7% in the VG versus 80.2% in the CG (P = 0.08). Due to trial design, 65% of patients received less than the optimal vaccine dose. Five-year DFS was 94.6% in optimally dosed patients (P = 0.05 versus the CG) and 87.1% in suboptimally dosed patients. A voluntary booster program was initiated, and among the 21 patients that were optimally boosted, there was only one recurrence (DFS = 95.2%). CONCLUSION The E75 vaccine is safe and appears to have clinical efficacy. A phase III trial evaluating the optimal dose and including booster inoculations has been initiated. CLINICAL TRIALS NCT00841399, NCT00584789.
Collapse
Affiliation(s)
- E A Mittendorf
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - G T Clifton
- Blanchfield Army Community Hospital, Fort Campbell
| | - J P Holmes
- Redwood Regional Medical Group, Santa Rosa
| | - E Schneble
- Department of Surgery, Brooke Army Medical Center, Ft Sam Houston
| | - D van Echo
- Department of Hematology Oncology, Walter Reed Army Medical Center, Washington
| | - S Ponniah
- Department of Surgery, Cancer Vaccine Development Program, United States Military Cancer Institute, Uniformed Services University of the Health Sciences, Bethesda, USA
| | - G E Peoples
- Department of Surgery, Brooke Army Medical Center, Ft Sam Houston; Department of Surgery, Cancer Vaccine Development Program, United States Military Cancer Institute, Uniformed Services University of the Health Sciences, Bethesda, USA.
| |
Collapse
|
78
|
Aspord C, Leloup C, Reche S, Plumas J. pDCs efficiently process synthetic long peptides to induce functional virus- and tumour-specific T-cell responses. Eur J Immunol 2014; 44:2880-92. [DOI: 10.1002/eji.201444588] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 06/12/2014] [Accepted: 07/04/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Caroline Aspord
- R&D Laboratory; Etablissement Français du Sang Rhone-Alpes; Grenoble France
- Immunobiology & Immunotherapy of Cancers; University Joseph Fourier; Grenoble France
| | - Claire Leloup
- R&D Laboratory; Etablissement Français du Sang Rhone-Alpes; Grenoble France
- Immunobiology & Immunotherapy of Cancers; University Joseph Fourier; Grenoble France
| | - Sabine Reche
- R&D Laboratory; Etablissement Français du Sang Rhone-Alpes; Grenoble France
- Immunobiology & Immunotherapy of Cancers; University Joseph Fourier; Grenoble France
| | - Joel Plumas
- R&D Laboratory; Etablissement Français du Sang Rhone-Alpes; Grenoble France
- Immunobiology & Immunotherapy of Cancers; University Joseph Fourier; Grenoble France
| |
Collapse
|
79
|
Abstract
Despite major advances in the treatment of ovarian cancer over the past two decades, it is still an incurable disease and requires the development of better treatment strategies. In recent years, we have developed a greater understanding of tumor immunology and the interactions between tumors and the immune system. This has led to the emergence of cancer immunotherapy as the fourth treatment modality in cancer. In this article, we address the principles of immunotherapy and different approaches that have been investigated over the past decade and discuss the future of immune therapy in ovarian cancer.
Collapse
Affiliation(s)
- Yousef Zakharia
- a Georgia Regents University Cancer Center, Augusta, Georgia; and
| | | | | |
Collapse
|
80
|
Abstract
The therapeutic potential of host-specific and tumour-specific immune responses is well recognized and, after many years, active immunotherapies directed at inducing or augmenting these responses are entering clinical practice. Antitumour immunization is a complex, multi-component task, and the optimal combinations of antigens, adjuvants, delivery vehicles and routes of administration are not yet identified. Active immunotherapy must also address the immunosuppressive and tolerogenic mechanisms deployed by tumours. This Review provides an overview of new results from clinical studies of therapeutic cancer vaccines directed against tumour-associated antigens and discusses their implications for the use of active immunotherapy.
Collapse
|
81
|
Occhipinti S, Sponton L, Rolla S, Caorsi C, Novarino A, Donadio M, Bustreo S, Satolli MA, Pecchioni C, Marchini C, Amici A, Cavallo F, Cappello P, Pierobon D, Novelli F, Giovarelli M. Chimeric rat/human HER2 efficiently circumvents HER2 tolerance in cancer patients. Clin Cancer Res 2014; 20:2910-21. [PMID: 24668647 DOI: 10.1158/1078-0432.ccr-13-2663] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite the great success of HER2 vaccine strategies in animal models, effective clinical results have not yet been obtained. We studied the feasibility of using DNA coding for chimeric rat/human HER2 as a tool to break the unresponsiveness of T cells from patients with HER2-overexpressing tumors (HER2-CP). EXPERIMENTAL DESIGN Dendritic cells (DCs) generated from patients with HER2-overexpressing breast (n = 28) and pancreatic (n = 16) cancer were transfected with DNA plasmids that express human HER2 or heterologous rat sequences in separate plasmids or as chimeric constructs encoding rat/human HER2 fusion proteins and used to activate autologous T cells. Activation was evaluated by IFN-γ ELISPOT assay, perforin expression, and ability to halt HER2+ tumor growth in vivo. RESULTS Specific sustained proliferation and IFN-γ production by CD4 and CD8 T cells from HER2-CP was observed after stimulation with autologous DCs transfected with chimeric rat/human HER2 plasmids. Instead, T cells from healthy donors (n = 22) could be easily stimulated with autologous DCs transfected with any human, rat, or chimeric rat/human HER2 plasmid. Chimeric HER2-transfected DCs from HER2-CP were also able to induce a sustained T-cell response that significantly hindered the in vivo growth of HER2(+) tumors. The efficacy of chimeric plasmids in overcoming tumor-induced T-cell dysfunction relies on their ability to circumvent suppressor effects exerted by regulatory T cells (Treg) and/or interleukin (IL)-10 and TGF-β1. CONCLUSIONS These results provide the proof of concept that chimeric rat/human HER2 plasmids can be used as effective vaccines for any HER2-CP with the advantage of being not limited to specific MHC. Clin Cancer Res; 20(11); 2910-21. ©2014 AACR.
Collapse
Affiliation(s)
- Sergio Occhipinti
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Laura Sponton
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Simona Rolla
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Cristiana Caorsi
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Anna Novarino
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Michela Donadio
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Sara Bustreo
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Maria Antonietta Satolli
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Carla Pecchioni
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Cristina Marchini
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Augusto Amici
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Federica Cavallo
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Paola Cappello
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Daniele Pierobon
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Francesco Novelli
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| | - Mirella Giovarelli
- Authors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, ItalyAuthors' Affiliations: Departments of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy; Center for Experimental Research and Medical Studies (CERMS), AO Città della Salute e della Scienza di Torino, Torino, Italy; Immunogenetic and Transplant Biology Service, AO Città della Salute e della Scienza Torino, Italy; Division of Oncology, Subalpine OncoHematology Cancer Center (COES), AO Città della Salute e della Scienza di Torino, Torino, Italy; Department of Oncology, University of Turin, Orbassano, Italy; Department of Medical Sciences, University of Torino, Torino, Italy, Department of Molecular Cellular and Animal Biology, University of Camerino, Camerino, Italy
| |
Collapse
|
82
|
Bollard CM, Gottschalk S, Torrano V, Diouf O, Ku S, Hazrat Y, Carrum G, Ramos C, Fayad L, Shpall EJ, Pro B, Liu H, Wu MF, Lee D, Sheehan AM, Zu Y, Gee AP, Brenner MK, Heslop HE, Rooney CM. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J Clin Oncol 2014; 32:798-808. [PMID: 24344220 PMCID: PMC3940538 DOI: 10.1200/jco.2013.51.5304] [Citation(s) in RCA: 405] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Tumor cells from approximately 40% of patients with Hodgkin or non-Hodgkin lymphoma express the type II latency Epstein-Barr virus (EBV) antigens latent membrane protein 1 (LMP1) and LMP2, which represent attractive targets for immunotherapy. Because T cells specific for these antigens are present with low frequency and may be rendered anergic by the tumors that express them, we expanded LMP-cytotoxic T lymphocytes (CTLs) from patients with lymphoma using autologous dendritic cells and EBV-transformed B-lymphoblastoid cell lines transduced with an adenoviral vector expressing either LMP2 alone (n = 17) or both LMP2 and ΔLMP1 (n = 33). PATIENTS AND METHODS These genetically modified antigen-presenting cells expanded CTLs that were enriched for specificity against type II latency LMP antigens. When infused into 50 patients with EBV-associated lymphoma, the expanded CTLs did not produce infusional toxicities. RESULTS Twenty-eight of 29 high-risk or multiple-relapse patients receiving LMP-CTLs as adjuvant therapy remained in remission at a median of 3.1 years after CTL infusion. None subsequently died as a result of lymphoma, but nine succumbed to complications associated with extensive prior chemoradiotherapy, including myocardial infarction and secondary malignancies. Of 21 patients with relapsed or resistant disease at the time of CTL infusion, 13 had clinical responses, including 11 complete responses. T cells specific for LMP as well as nonviral tumor-associated antigens (epitope spreading) could be detected in the peripheral blood within 2 months after CTL infusion, but this evidence for epitope spreading was seen only in patients achieving clinical responses. CONCLUSION Autologous T cells directed to the LMP2 or LMP1 and LMP2 antigens can induce durable complete responses without significant toxicity. Their earlier use in the disease course may reduce delayed treatment-related mortality.
Collapse
Affiliation(s)
- Catherine M. Bollard
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Stephen Gottschalk
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Vicky Torrano
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Oumar Diouf
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Stephanie Ku
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Yasmin Hazrat
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - George Carrum
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Carlos Ramos
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Luis Fayad
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Elizabeth J. Shpall
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Barbara Pro
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Hao Liu
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Meng-Fen Wu
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Daniel Lee
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Andrea M. Sheehan
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Youli Zu
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Adrian P. Gee
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Malcolm K. Brenner
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Helen E. Heslop
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| | - Cliona M. Rooney
- Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Andrea M. Sheehan, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Baylor College of Medicine and Texas Children's Hospital; Catherine M. Bollard, Stephen Gottschalk, Vicky Torrano, Oumar Diouf, Stephanie Ku, Yasmin Hazrat, George Carrum, Carlos Ramos, Hao Liu, Meng-Fen Wu, Daniel Lee, Andrea M. Sheehan, Youli Zu, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, and Cliona M. Rooney, Methodist Hospital; Luis Fayad, Elizabeth J. Shpall, and Barbara Pro, MD Anderson Cancer Center, Houston, TX; and Daniel Lee and Youli Zu, Weill Medical College of Cornell University, New York, NY
| |
Collapse
|
83
|
Curigliano G, Spitaleri G, Dettori M, Locatelli M, Scarano E, Goldhirsch A. Vaccine immunotherapy in breast cancer treatment: promising, but still early. Expert Rev Anticancer Ther 2014; 7:1225-41. [PMID: 17892423 DOI: 10.1586/14737140.7.9.1225] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer vaccine-based immunotherapy should potentiate immunosurveillance function, preventing and protecting against growing tumors. Tumor cells usually activate the immune system, including T lymphocytes and natural killer cells, which are able to eliminate the transformed cells. Immunosubversion mechanisms related to tumor cells antigenic immunoediting induces mechanisms of tolerance and immunoescape. This condition impairs not only host-generated immunosurveillance, but also attempts to harness the immune response for therapeutic purposes. Most trials evaluating breast cancer vaccines have been carried out in patients in the metastatic and adjuvant setting. The aim of this review is to analyze the activity of vaccination strategies in current clinical trials. We summarize the differential approaches, protein-based and cell-based vaccines, focusing on vaccines targeting HER2/neu protein. Another focus of the review is to provide the reader with future challenges in the field, taking into account both the immunological and clinical aspects to better target the goal.
Collapse
Affiliation(s)
- Giuseppe Curigliano
- European Institute of Oncology, Department of Medicine, Division of Medical Oncology, Via Ripamonti 435, 20141 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
84
|
Abstract
Ovarian cancer is the leading cause of gynecologic cancer deaths and accounts for 4% of women's cancer diagnoses and 5% of all cancer mortalities. Despite the ability of current chemotherapy and cytoreductive surgery to put patients in remission, most patients with advanced cancer will eventually relapse. Many advances in the treatment of ovarian cancer have been reported in the past several years and a historical background is provided. Attention will then turn to analogs of current chemotherapeutic agents, new cytotoxic drugs, targeted molecular therapy, intraperitoneal therapy and immunotherapy. This review will give a perspective on current drugs, potential agents and upcoming clinical trials.
Collapse
Affiliation(s)
- Franco Muggia
- New York University Clinical Cancer Center, NY 10016-9196, USA.
| | | |
Collapse
|
85
|
Tagliamonte M, Petrizzo A, Tornesello ML, Buonaguro FM, Buonaguro L. Antigen-specific vaccines for cancer treatment. Hum Vaccin Immunother 2014; 10:3332-3346. [PMID: 25483639 PMCID: PMC4514024 DOI: 10.4161/21645515.2014.973317] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 08/08/2014] [Accepted: 08/23/2014] [Indexed: 12/14/2022] Open
Abstract
Vaccines targeting pathogens are generally effective and protective because based on foreign non-self antigens which are extremely potent in eliciting an immune response. On the contrary, efficacy of therapeutic cancer vaccines is still disappointing. One of the major reasons for such poor outcome, among others, is the difficulty of identifying tumor-specific target antigens which should be unique to the tumors or, at least, overexpressed on the tumors as compared to normal cells. Indeed, this is the only option to overcome the peripheral immune tolerance and elicit a non toxic immune response. New and more potent strategies are now available to identify specific tumor-associated antigens for development of cancer vaccine approaches aiming at eliciting targeted anti-tumor cellular responses. In the last years this aspect has been addressed and many therapeutic vaccination strategies based on either whole tumor cells or specific antigens have been and are being currently evaluated in clinical trials. This review summarizes the current state of cancer vaccines, mainly focusing on antigen-specific approaches.
Collapse
Key Words
- APCs, antigen-presenting cell
- BCG, Bacille Calmette-Guerin
- BCR, B-cell receptor
- CDCA1, cell division cycle associated 1
- CRC, colorectal cancer
- CT, Cancer-testis
- CTL, cytotoxic T-lympocites
- DCs, dendritic cells
- EGT, electro-gene-transfer
- FDA, Food & drug administration
- GB, glioblastoma
- GM-CSF, granulocyte macrophage-colony stimulating factor
- HER2, human epidermal growth factor receptor 2
- HLA, human leukocyte antigen
- HPV, human papillomavirus
- HSPs, stress/heat shock proteins
- IFNg, interferon gamma
- Ig Id, immunoglobulin idiotype
- LPs, long peptides
- MAGE-A1, Melanoma-associated antigen 1
- MHC, major histocompatibility complex
- MS, mass spectrometry
- MVA, modified vaccinia strain Ankara
- NSCLC, non-small-cell lung carcinoma
- PAP, prostatic acid phosphatase
- PRRs, Pattern Recognition Receptors
- PSA, Prostate-specific antigen
- RCR, renal cell cancer
- SSX-2, Synovial sarcoma X breakpoint 2
- TAAs, tumor-associated antigens
- TACAs, Tumor-associated carbohydrate antigens
- TARP, T-cell receptor gamma alternate reading frame protein
- TLRs, Toll-Like Receptors
- TPA, transporter associated with antigen processing
- WES, whole exome sequencing
- WGS, whole genome sequencing
- cancer vaccine
- clinical trials
- epitopes
- hTERT, human Telomerase reverse transcriptase
- immunotherapeutics
- mCRPC, metastatic castrate-resistant prostate cancer
- tumor-associated antigens
Collapse
Affiliation(s)
- Maria Tagliamonte
- Laboratory of Molecular Biology and Viral Oncology; Department of Experimental Oncology; Istituto Nazionale per lo Studio e la Cura dei Tumori; “Fondazione Pascale” - IRCCS; Naples, Italy
| | - Annacarmen Petrizzo
- Laboratory of Molecular Biology and Viral Oncology; Department of Experimental Oncology; Istituto Nazionale per lo Studio e la Cura dei Tumori; “Fondazione Pascale” - IRCCS; Naples, Italy
| | - Maria Lina Tornesello
- Laboratory of Molecular Biology and Viral Oncology; Department of Experimental Oncology; Istituto Nazionale per lo Studio e la Cura dei Tumori; “Fondazione Pascale” - IRCCS; Naples, Italy
| | - Franco M Buonaguro
- Laboratory of Molecular Biology and Viral Oncology; Department of Experimental Oncology; Istituto Nazionale per lo Studio e la Cura dei Tumori; “Fondazione Pascale” - IRCCS; Naples, Italy
| | - Luigi Buonaguro
- Laboratory of Molecular Biology and Viral Oncology; Department of Experimental Oncology; Istituto Nazionale per lo Studio e la Cura dei Tumori; “Fondazione Pascale” - IRCCS; Naples, Italy
| |
Collapse
|
86
|
Gottschalk S, Yu F, Ji M, Kakarla S, Song XT. A vaccine that co-targets tumor cells and cancer associated fibroblasts results in enhanced antitumor activity by inducing antigen spreading. PLoS One 2013; 8:e82658. [PMID: 24349329 PMCID: PMC3861387 DOI: 10.1371/journal.pone.0082658] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/25/2013] [Indexed: 11/18/2022] Open
Abstract
Dendritic cell (DC) vaccines targeting only cancer cells have produced limited antitumor activity in most clinical studies. Targeting cancer-associated fibroblasts (CAFs) in addition to cancer cells may enhance antitumor effects, since CAFs, the central component of the tumor stroma, directly support tumor growth and contribute to the immunosuppressive tumor microenvironment. To co-target CAFs and tumor cells we developed a new compound DC vaccine that encodes an A20-specific shRNA to enhance DC function, and targets fibroblast activation protein (FAP) expressed in CAFs and the tumor antigen tyrosine-related protein (TRP)2 (DC-shA20-FAP-TRP2). DC-shA20-FAP-TRP2 vaccination induced robust FAP- and TRP2-specific T-cell responses, resulting in greater antitumor activity in the B16 melanoma model in comparison to monovalent vaccines or a vaccine encoding antigens and a control shRNA. DC-shA20-FAP-TRP2 vaccination enhanced tumor infiltration of CD8-positive T cells, and induced antigen-spreading resulting in potent antitumor activity. Thus, co-targeting of tumor cells and CAFs results in the induction of broad-based tumor-specific T-cell responses and has the potential to improve current vaccine approaches for cancer.
Collapse
Affiliation(s)
- Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (XTS); (SG)
| | - Feng Yu
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Minjun Ji
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sunitha Kakarla
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xiao-Tong Song
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (XTS); (SG)
| |
Collapse
|
87
|
Disis ML, Gad E, Herendeen DR, Lai VP, Park KH, Cecil DL, O'Meara MM, Treuting PM, Lubet RA. A multiantigen vaccine targeting neu, IGFBP-2, and IGF-IR prevents tumor progression in mice with preinvasive breast disease. Cancer Prev Res (Phila) 2013; 6:1273-82. [PMID: 24154719 PMCID: PMC3864759 DOI: 10.1158/1940-6207.capr-13-0182] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A multiantigen multipeptide vaccine, targeting proteins expressed in preinvasive breast lesions, can stimulate type I CD4(+) T cells which have been shown to be deficient in both patients with breast cancer and mice that develop mammary tumors. Transgenic mice (TgMMTV-neu) were immunized with a multiantigen peptide vaccine specific for neu, insulin-like growth factor-binding protein 2 and insulin-like growth factor receptor-I at a time when some of the animals already had preinvasive lesions (18 weeks of age). Although immunization with each individual antigen was partially effective in inhibiting tumor growth, immunization with the multiantigen vaccine was highly effective, blocking development of palpable lesions in 65% of mice and slowing tumor growth in the infrequent palpable tumors, which did arise. Protection was mediated by CD4(+) T cells, and the few slow-growing tumors that did develop demonstrated a significant increase in intratumoral CD8(+) T cells as compared with controls (P = 0.0007). We also combined the vaccine with agents that were, by themselves, partially effective inhibitors of tumor progression in this model; lapatinib and the RXR agonist bexarotene. Although the combination of lapatinib and vaccination performed similarly to vaccination alone (P = 0.735), bexarotene and vaccination significantly enhanced disease-free survival (P < 0.0001), and approximately 90% of the mice showed no pathologic evidence of carcinomas at one year. The vaccine also demonstrated significant clinical efficacy in an additional transgenic model of breast cancer (TgC3(I)-Tag). Chemoimmunoprevention combinations may be an effective approach to breast cancer prevention even when the vaccine is administered in the presence of subclinical disease.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Bexarotene
- Female
- Insulin-Like Growth Factor Binding Protein 2/antagonists & inhibitors
- Insulin-Like Growth Factor Binding Protein 2/immunology
- Lapatinib
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/pathology
- Mammary Neoplasms, Animal/prevention & control
- Mice
- Mice, Transgenic
- Neoplasm Invasiveness
- Precancerous Conditions/immunology
- Precancerous Conditions/pathology
- Precancerous Conditions/prevention & control
- Quinazolines/administration & dosage
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/immunology
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Tetrahydronaphthalenes/administration & dosage
- Tumor Cells, Cultured
- Vaccines, Subunit/therapeutic use
Collapse
Affiliation(s)
- Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, 850 Republican Street, Box 358050, University of Washington, Seattle, WA 98109.
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Lavoué V, Foucher F, Henno S, Bauville E, Catros V, Cabillic F, Levêque J. [Immunotherapy in epithelial ovarian carcinoma: hope and reality]. ACTA ACUST UNITED AC 2013; 43:198-210. [PMID: 24230482 DOI: 10.1016/j.jgyn.2013.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/14/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Epithelial ovarian carcinoma (EOC) has a worst prognosis with little progress in terms of survival for the last two decades. Immunology received little interest in EOC in the past, but now appears very important in the natural history of this cancer. This review is an EOC immunology state of art and focuses on the place of immunotherapy in future. MATERIAL AND METHODS A systematic review of published studies was performed. Medline baseline interrogation was performed with the following keywords: "Ovarian carinoma, immunotherapy, T-lymphocyte, regulator T-lymphocyte, dendritic cells, macrophage, antigen, chemotherapy, surgery, clinical trials". Identified publications (English or French) were assessed for the understanding of EOC immunology and the place of conventional treatment and immunotherapy strategy. RESULTS Intratumoral infiltration by immune cells is a strong prognotic factor in EOC. Surgery and chemotherapy in EOC decrease imunosuppression in patients. The antitumoral immunity is a part of the therapeutic action of surgery and chemotherapy. Until now, immunotherapy gave some disappointing results, but the new drugs that target the tolerogenic tumoral microenvironnement rise and give a new hope in the treatment of cancer. CONCLUSION Immunology controls the EOC natural history. The modulation of immunosuppressive microenvironment associated with the stimulation of antitumoral immunity could be the next revolution in the treatment of cancer.
Collapse
Affiliation(s)
- V Lavoué
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France; Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France.
| | - F Foucher
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - S Henno
- Service d'anatomo-pathologie, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - E Bauville
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - V Catros
- Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France; Service de biologie cellulaire, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - F Cabillic
- Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France; Service de biologie cellulaire, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - J Levêque
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France; Faculté de médecine, université de Rennes 1, 35000 Rennes, France
| |
Collapse
|
89
|
Disis ML, Dang Y, Coveler AL, Marzbani E, Kou ZC, Childs JS, Fintak P, Higgins DM, Reichow J, Waisman J, Salazar LG. HER-2/neu vaccine-primed autologous T-cell infusions for the treatment of advanced stage HER-2/neu expressing cancers. Cancer Immunol Immunother 2013; 63:101-9. [PMID: 24162107 DOI: 10.1007/s00262-013-1489-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 10/12/2013] [Indexed: 12/31/2022]
Abstract
This phase I study evaluated the feasibility of expanding HER-2/neu (HER2) vaccine-primed peripheral blood T-cells ex vivo and assessed the safety of T-cell infusions. Eight patients with HER2(+) treatment refractory metastatic cancers were enrolled. T-cells could be expanded to predefined parameters in seven patients (88%). Ninety-two percent of adverse events were grade 1 or 2. Three of seven patients developed infusion-related inflammatory reactions at their disease sites. HER2-specific T-cells significantly increased in vivo compared to pre-infusion levels (p = 0.010) and persisted in 4/6 patients (66%) over 70 days after the first infusion. Partial clinical responses were observed in 43% of patients. Levels of T-regulatory cells in peripheral blood prior to infusion (p < 0.001), the level of HER2-specific T-cells in vivo (p = 0.030), and development of diverse clonal T-cell populations (p < 0.001) were associated with response. The generation of HER2 vaccine-primed autologous T-cells for therapeutic infusion is feasible and well tolerated. This approach provides a foundation for the application of T-cell therapy to additional solid tumor types.
Collapse
Affiliation(s)
- Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, 850 Republican Street, Box 358050, Seattle, WA, 98195-8050, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
DNA vaccines targeting the encoded antigens to dendritic cells induce potent antitumor immunity in mice. BMC Immunol 2013; 14:39. [PMID: 23941509 PMCID: PMC3751307 DOI: 10.1186/1471-2172-14-39] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 07/31/2013] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Although DNA vaccine holds a great potential for cancer immunotherapy, effective long-lasting antitumoral immunity sufficient to induce durable responses in cancer patients remains to be achieved. Considering the pivotal role of dendritic cells (DC) in the antigen processing and presentation, we prepared DC-targeting DNA vaccines by fusing tumor-associated antigen HER2/neu ectodomain to single chain antibody fragment (scFv) from NLDC-145 antibody specific for DC-restricted surface molecule DEC-205 (scFvNLDC-145), and explored its antitumoral efficacy and underlying mechanisms in mouse breast cancer models. RESULTS In vivo targeting assay demonstrated that scFvNLDC-145 specifically delivered DNA vaccine-encoded antigen to DC. Compared with untargeted HER2/neu DNA vaccines, vaccination with scFvNLDC-145-HER2/neu markedly promoted the HER2/neu-specific cellular and humoral immune responses with long-lasting immune memory, resulting in effective protection against challenge of HER2/neu-positive D2F2/E2 breast tumor while ineffective in parental HER2/neu-negative D2F2 breast tumor. More importantly, in combination with temporary depletion of regulatory T cells (Treg) by low-dose cyclophosphamide, vaccination with scFvNLDC-145-HER2/neu induced the regression of established D2F2/E2 breast tumor and significantly retarded the development of spontaneous mammary carcinomas in transgenic BALB-neuT mice. CONCLUSION Our findings demonstrate that DC-targeted DNA vaccines for in vivo direct delivery of tumor antigens to DC could induce potent antigen-specific cellular and humoral immune responses and, if additional combination with systemic Treg depletion, was able to elicit an impressively therapeutic antitumoral activity, providing a rationale for further development of this approach for cancer treatment.
Collapse
|
91
|
Milani A, Sangiolo D, Montemurro F, Aglietta M, Valabrega G. Active immunotherapy in HER2 overexpressing breast cancer: current status and future perspectives. Ann Oncol 2013; 24:1740-1748. [PMID: 23585514 DOI: 10.1093/annonc/mdt133] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND The use of anti-HER2 monoclonal antibodies (mAbs) has improved the clinical outcome of HER2-overexpressing breast cancers (BCs). Unfortunately, often these tumors tend to relapse and, when metastatic, the duration of clinical benefit is limited over time and almost invariably followed by tumor progression. Alternative approaches to this essentially passive immunotherapy are therefore needed in HER2-overexpressing BC patients. As HER2 is one of the most suitable targets for active immunotherapy in BC, manipulating the immune system is a highly attractive approach. MATERIAL AND METHODS A computer-based literature search was carried out using PubMed (keywords: breast neoplasm, HER2 vaccine, immunology); data reported at international meetings were included. RESULTS This review provides a focus on the following active vaccinal approaches under clinical investigation against HER2-overexpressing BC: (i) peptide and protein based; (ii) DNA based; (iii) whole tumor cell based; (iv) dendritic cell based. Moreover, the review discuss future challenges in the field, trying to define the best setting for the development of this innovative strategy, considering both immunological and clinical aspects of HER2 targeting. CONCLUSIONS Development of effective vaccines for BC remains a distinct challenge but is likely to become a substantial advance for patients with HER2-overexpressing BCs.
Collapse
Affiliation(s)
- A Milani
- Medical Oncology I, Institute for Cancer Research and Treatment (IRCC) Candiolo FPO (Fondazione del Piemonte per l'Oncologia); Department of Oncology, University of Torino Medical School, Candiolo
| | - D Sangiolo
- Medical Oncology I, Institute for Cancer Research and Treatment (IRCC) Candiolo FPO (Fondazione del Piemonte per l'Oncologia); Department of Oncology, University of Torino Medical School, Candiolo
| | - F Montemurro
- Medical Oncology I, Institute for Cancer Research and Treatment (IRCC) Candiolo FPO (Fondazione del Piemonte per l'Oncologia); Investigative Clinical Oncology Unit (INCO), Candiolo, Italy
| | - M Aglietta
- Medical Oncology I, Institute for Cancer Research and Treatment (IRCC) Candiolo FPO (Fondazione del Piemonte per l'Oncologia); Department of Oncology, University of Torino Medical School, Candiolo
| | - G Valabrega
- Medical Oncology I, Institute for Cancer Research and Treatment (IRCC) Candiolo FPO (Fondazione del Piemonte per l'Oncologia); Department of Oncology, University of Torino Medical School, Candiolo.
| |
Collapse
|
92
|
Broussard EK, Kim R, Wiley JC, Marquez JP, Annis JE, Pritchard D, Disis ML. Identification of putative immunologic targets for colon cancer prevention based on conserved gene upregulation from preinvasive to malignant lesions. Cancer Prev Res (Phila) 2013; 6:666-74. [PMID: 23682078 PMCID: PMC3718634 DOI: 10.1158/1940-6207.capr-12-0484] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The length of time required for preinvasive adenoma to progress to carcinoma, the immunogenicity of colorectal cancer (CRC), and the identification of high-risk populations make development and testing of a prophylactic vaccine for the prevention of CRC possible. We hypothesized that genes upregulated in adenoma relative to normal tissue, which maintained increased expression in CRC, would encode proteins suitable as putative targets for immunoprevention. We evaluated existing adenoma and CRC microarray datasets and identified 160 genes that were ≥2-fold upregulated in both adenoma and CRC relative to normal colon tissue. We further identified 23 genes that showed protein overexpression in colon adenoma and CRC based on literature review. Silencing the most highly upregulated genes, CDH3, CLDN1, KRT23, and MMP7, in adenoma and CRC cell lines resulted in a significant decrease in viability (P < 0.0001) and proliferation (P < 0.0001) as compared to controls and an increase in cellular apoptosis (P < 0.05 for CDH3, KRT23). Results were duplicated across cell lines representing microsatellite instability, CpG island methylator, and chromosomal instability phenotypes, suggesting immunologic elimination of cells expressing these proteins could impact the progression of all CRC phenotypes. To determine whether these proteins were immunogens, we interrogated sera from early stage CRC patients and controls and found significantly elevated CDH3 (P = 0.006), KRT23 (P = 0.0007), and MMP7 (P < 0.0001) serum immunoglobulin G in cases as compared to controls. These data show a high throughput approach to the identification of biologically relevant putative immunologic targets for CRC and identified three candidates suitable for vaccine development.
Collapse
MESH Headings
- Adenoma/diagnosis
- Adenoma/metabolism
- Adenoma/prevention & control
- Adult
- Aged
- Aged, 80 and over
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cadherins/antagonists & inhibitors
- Cadherins/genetics
- Cadherins/metabolism
- Case-Control Studies
- Cell Proliferation
- Claudin-1/antagonists & inhibitors
- Claudin-1/genetics
- Claudin-1/metabolism
- Colorectal Neoplasms/diagnosis
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/prevention & control
- DNA Methylation
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Keratins, Type I/antagonists & inhibitors
- Keratins, Type I/genetics
- Keratins, Type I/metabolism
- Male
- Matrix Metalloproteinase 7/chemistry
- Matrix Metalloproteinase 7/genetics
- Matrix Metalloproteinase 7/metabolism
- Microsatellite Instability
- Middle Aged
- Neoplasm Staging
- Precancerous Conditions/diagnosis
- Precancerous Conditions/metabolism
- Precancerous Conditions/prevention & control
- Prognosis
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Young Adult
Collapse
Affiliation(s)
- Elizabeth K Broussard
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA 98109, USA.
| | | | | | | | | | | | | |
Collapse
|
93
|
Abstract
While therapeutic vaccines for ovarian cancer represent only a small fraction of active clinical trials, growing interest in this area and the accumulated data supporting the use of vaccines in cancer treatment portend further expansion of trials incorporating these strategies. This review explores the rationale for the use of vaccines for the treatment of ovarian cancer. It examines vaccine platforms that have been investigated and reviews the data from these studies. We also highlight recently reported phase 2 and 3 clinical trials with clinical outcomes as endpoints. Finally, we consider directions for the next generation of vaccines in light of these findings and our emerging understanding of agents that may augment vaccine responses by targeting the immunosuppressive impact of the tumor microenvironment.
Collapse
|
94
|
Lavoué V, Thédrez A, Levêque J, Foucher F, Henno S, Jauffret V, Belaud-Rotureau MA, Catros V, Cabillic F. Immunity of human epithelial ovarian carcinoma: the paradigm of immune suppression in cancer. J Transl Med 2013; 11:147. [PMID: 23763830 PMCID: PMC3683338 DOI: 10.1186/1479-5876-11-147] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 05/27/2013] [Indexed: 12/21/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a significant cause of cancer-related mortality in women, and there has been no substantial decrease in the death rates due to EOC in the last three decades. Thus, basic knowledge regarding ovarian tumor cell biology is urgently needed to allow the development of innovative treatments for EOC. Traditionally, EOC has not been considered an immunogenic tumor, but there is evidence of an immune response to EOC in patients. Clinical data demonstrate that an antitumor immune response and immune evasion mechanisms are correlated with a better and lower survival, respectively, providing evidence for the immunoediting hypothesis in EOC. This review focuses on the immune response and immune suppression in EOC. The immunological roles of chemotherapy and surgery in EOC are also described. Finally, we detail pilot data supporting the efficiency of immunotherapy in the treatment of EOC and the emerging concept that immunomodulation aimed at counteracting the immunosuppressive microenvironment must be associated with immunotherapy strategies.
Collapse
Affiliation(s)
- Vincent Lavoué
- Lady Davis Institut, Jewish General Hospital, McGill University, Montreal QC H3T 1E2, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Spear P, Barber A, Rynda-Apple A, Sentman CL. NKG2D CAR T-cell therapy inhibits the growth of NKG2D ligand heterogeneous tumors. Immunol Cell Biol 2013; 91:435-40. [PMID: 23628805 DOI: 10.1038/icb.2013.17] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 03/21/2013] [Accepted: 03/21/2013] [Indexed: 12/12/2022]
Abstract
Tumor heterogeneity presents a substantial barrier to increasing clinical responses mediated by targeted therapies. Broadening the immune response elicited by treatments that target a single antigen is necessary for the elimination of tumor variants that fail to express the targeted antigen. In this study, it is shown that adoptive transfer of T cells bearing a chimeric antigen receptor (CAR) inhibited the growth of target-expressing and -deficient tumor cells within ovarian and lymphoma tumors. Mice bearing the ID8 ovarian or RMA lymphoma tumors were treated with T cells transduced with a NKG2D-based CAR (chNKG2D). NKG2D CAR T-cell therapy protected mice from heterogeneous RMA tumors. Moreover, adoptive transfer of chNKG2D T cells mediated tumor protection against highly heterogeneous ovarian tumors in which 50, 20 or only 7% of tumor cells expressed significant amounts of NKG2D ligands. CAR T cells did not mediate an in vivo response against tumor cells that did not express sufficient amounts of NKG2D ligands, and the number of ligand-expressing tumor cells correlated with therapeutic efficacy. In addition, tumor-free surviving mice were protected against a tumor re-challenge with NKG2D ligand-negative ovarian tumor cells. These data indicate that NKG2D CAR T-cell treatment can be an effective therapy against heterogeneous tumors and induce tumor-specific immunity against ligand-deficient tumor cells.
Collapse
Affiliation(s)
- Paul Spear
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | | | | |
Collapse
|
96
|
Vaccination for the prevention and treatment of breast cancer with special focus on Her-2/neu peptide vaccines. Breast Cancer Res Treat 2013; 138:1-12. [PMID: 23340862 DOI: 10.1007/s10549-013-2410-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 01/07/2013] [Indexed: 01/18/2023]
Abstract
Immunologic interventions in a subset of breast cancer patients represent a well-established therapeutic approach reflecting individualized treatment modalities. Thus, the therapeutic administration of monoclonal antibodies targeting tumor-associated antigens (TAA), such as Her-2/neu, represents a milestone in cancer treatment. However, passive antibody administration suffers from several drawbacks, including frequency and long duration of treatment. These undesirables may be avoidable in an approach based on generating active immune responses against these same targets. Only recently has the significance of tumors in relation to their microenvironments been understood as essential for creating an effective cancer vaccine. In particular, the immune system plays an important role in suppressing or promoting tumor formation and growth. Therefore, activation of appropriate triggers (such as induction of Th1 cells, CD8+ T cells, and suppression of regulatory cells in combination with generation of antibodies with anti-tumor activity) is a desirable goal. Current vaccination approaches have concentrated on therapeutic vaccines using certain TAA. Many cancer antigens, including breast cancer antigens, have been described and also given priority ranking for use as vaccine antigens by the US National Cancer Institute. One of the TAA antigens which has been thoroughly examined in numerous trials is Her-2/neu. This review will discuss delivery systems for this antigen with special focus on T and B cell peptide vaccines. Attention will be given to their advantages and limitations, as well as the use of certain adjuvants to improve anti-cancer responses.
Collapse
|
97
|
Patronov A, Doytchinova I. T-cell epitope vaccine design by immunoinformatics. Open Biol 2013; 3:120139. [PMID: 23303307 PMCID: PMC3603454 DOI: 10.1098/rsob.120139] [Citation(s) in RCA: 266] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Accepted: 12/11/2012] [Indexed: 01/08/2023] Open
Abstract
Vaccination is generally considered to be the most effective method of preventing infectious diseases. All vaccinations work by presenting a foreign antigen to the immune system in order to evoke an immune response. The active agent of a vaccine may be intact but inactivated ('attenuated') forms of the causative pathogens (bacteria or viruses), or purified components of the pathogen that have been found to be highly immunogenic. The increased understanding of antigen recognition at molecular level has resulted in the development of rationally designed peptide vaccines. The concept of peptide vaccines is based on identification and chemical synthesis of B-cell and T-cell epitopes which are immunodominant and can induce specific immune responses. The accelerating growth of bioinformatics techniques and applications along with the substantial amount of experimental data has given rise to a new field, called immunoinformatics. Immunoinformatics is a branch of bioinformatics dealing with in silico analysis and modelling of immunological data and problems. Different sequence- and structure-based immunoinformatics methods are reviewed in the paper.
Collapse
Affiliation(s)
| | - Irini Doytchinova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
98
|
Sunay M, Marincola F, Khleif SN, Silverstein SC, Fox BA, Galon J, Emens LA. Focus on the target: the tumor microenvironment, Society for Immunotherapy of Cancer Annual Meeting Workshop, October 24th-25th 2012. J Immunother Cancer 2013. [PMCID: PMC4019899 DOI: 10.1186/2051-1426-1-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Workshop associated with the 27th Annual Meeting of the Society for Immunotherapy of Cancer (SITC), North Bethesda, MD, October 24-25, 2012 focused on targeting the tumor microenvironment as part of an integrative approach to immune-based cancer therapy.
Collapse
|
99
|
Morse MA, Osada T, Hobeika A, Patel S, Lyerly HK. Biomarkers and correlative endpoints for immunotherapy trials. Am Soc Clin Oncol Educ Book 2013:0011300287. [PMID: 23714525 DOI: 10.14694/edbook_am.2013.33.e287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Immunotherapies for lung cancer are reaching phase III clinical trial, but the ultimate success likely will depend on developing biomarkers to guide development and choosing patient populations most likely to benefit. Because the immune response to cancer involves multiple cell types and cytokines, some spatially and temporally separated, it is likely that multiple biomarkers will be required to fully characterize efficacy of the vaccine and predict eventual benefit. Peripheral blood markers of response, such as the ELISPOT assay and cytokine flow cytometry analyses of peripheral blood mononuclear cells following immunotherapy, remain the standard approach, but it is increasingly important to obtain tissue to study the immune response at the site of the tumor. Earlier clinical endpoints such as response rate and progression-free survival do not correlate with overall survival demonstrated for some immunotherapies, suggesting the need to develop other intermediary clinical endpoints. Insofar as all these biomarkers and surrogate endpoints are relevant in multiple malignancies, it may be possible to extrapolate findings to immunotherapy of lung cancer.
Collapse
|
100
|
Hale DF, Clifton GT, Sears AK, Vreeland TJ, Shumway N, Peoples GE, Mittendorf EA. Cancer vaccines: should we be targeting patients with less aggressive disease? Expert Rev Vaccines 2012; 11:721-31. [PMID: 22873128 DOI: 10.1586/erv.12.39] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is enthusiasm for using vaccines to stimulate the immune system to treat cancer. In this article, the authors review the evolution of vaccines evaluated in clinical trials, starting with Phase III trials in metastatic disease and progressing to trials in the adjuvant setting. Data from these trials suggest that cancer vaccines may be more effective in patients with lower volume disease, and data from the E75 peptide vaccine trials suggest that vaccines may be most effective in less aggressive disease.
Collapse
Affiliation(s)
- Diane F Hale
- Department of Surgery, General Surgery Service, Brooke Army Medical Center, San Antonio, TX, USA
| | | | | | | | | | | | | |
Collapse
|