1
|
Telomerase-based GX301 cancer vaccine in patients with metastatic castration-resistant prostate cancer: a randomized phase II trial. Cancer Immunol Immunother 2021; 70:3679-3692. [PMID: 34351436 PMCID: PMC8571235 DOI: 10.1007/s00262-021-03024-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/25/2021] [Indexed: 12/19/2022]
Abstract
Debate is around the optimal immunization regimen for cancer vaccines since too intense vaccination schedules may exhaust reactive lymphocytes. GX301 is a telomerase-based cancer vaccine whose safety and immunological effects were tested in a phase I trial applying an eight administrations schedule. Main objective of this study was to comparatively analyse safety and immunological response to three GX301 regimens in metastatic castration-resistant prostate cancer patients with response/disease stability after docetaxel chemotherapy. This was a multicentre, randomized, parallel-group, open-label trial registered with EudraCT (2014-000095-26) and ClinicalTrials.gov (NCT02293707, 2014). Ninety-eight patients were randomized to receive either eight (regimen 1), four (regimen 2) or two (regimen 3) vaccine administrations. Sixty-three patients were assessable for the primary immunological end-point. Vaccine-specific immune responses were evaluated by intracellular staining for IFN, elispot and cytotoxic assay at 90 and 180 days from baseline. No major side effects were recorded. A 54% overall immune responder rate was observed with 95% of patients showing at least one vaccine-specific immune response. Rate of immunological responders and number of immunizations were proportionally related, suggesting superiority of regimens 1 and 2 over regimen 3. Overall survival did not differ among regimens in both immunological responders and non-responders and was inversely associated (P = 0.002) with increase in the number of circulating CD8 + T regulatory cells at 180 days. These data indicate that GX301 cancer vaccine is safe and immunogenic in metastatic castration-resistant prostate cancer patients. Schedules with high number of administrations should be preferred in future studies due to their better immunological outcome.
Collapse
|
2
|
Cheng R, Fontana F, Xiao J, Liu Z, Figueiredo P, Shahbazi MA, Wang S, Jin J, Torrieri G, Hirvonen JT, Zhang H, Chen T, Cui W, Lu Y, Santos HA. Recombination Monophosphoryl Lipid A-Derived Vacosome for the Development of Preventive Cancer Vaccines. ACS APPLIED MATERIALS & INTERFACES 2020; 12:44554-44562. [PMID: 32960566 PMCID: PMC7549091 DOI: 10.1021/acsami.0c15057] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 05/09/2023]
Abstract
Recently, there has been an increasing interest for utilizing the host immune system to fight against cancer. Moreover, cancer vaccines, which can stimulate the host immune system to respond to cancer in the long term, are being investigated as a promising approach to induce tumor-specific immunity. In this work, we prepared an effective cancer vaccine (denoted as "vacosome") by reconstructing the cancer cell membrane, monophosphoryl lipid A as a toll-like receptor 4 agonist, and egg phosphatidylcholine. The vacosome triggered and enhanced bone marrow dendritic cell maturation as well as stimulated the antitumor response against breast cancer 4T1 cells in vitro. Furthermore, an immune memory was established in BALB/c mice after three-time preimmunization with the vacosome. After that, the immunized mice showed inhibited tumor growth and prolonged survival period (longer than 50 days). Overall, our results demonstrate that the vacosome can be a potential candidate for clinical translation as a cancer vaccine.
Collapse
Affiliation(s)
- Ruoyu Cheng
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Flavia Fontana
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Junyuan Xiao
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Zehua Liu
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Patrícia Figueiredo
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mohammad-Ali Shahbazi
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran
| | - Shiqi Wang
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jing Jin
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Giulia Torrieri
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jouni T. Hirvonen
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Hongbo Zhang
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
- Department of Pharmaceutical Sciences Laboratory and
Turku Center for Biotechnology, Åbo
Akademi University, FI-20520 Turku, Finland
| | - Tongtong Chen
- Radiology Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, PR China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Yong Lu
- Radiology Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, PR China
| | - Hélder A. Santos
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
- Helsinki Insititute of Life Science, HiLIFE, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
3
|
Regulation of Murine Ovarian Epithelial Carcinoma by Vaccination against the Cytoplasmic Domain of Anti-Müllerian Hormone Receptor II. J Immunol Res 2015; 2015:630287. [PMID: 26618181 PMCID: PMC4651663 DOI: 10.1155/2015/630287] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/12/2015] [Indexed: 12/23/2022] Open
Abstract
Anti-Müllerian hormone receptor, type II (AMHR2), is a differentiation protein expressed in 90% of primary epithelial ovarian carcinomas (EOCs), the most deadly gynecologic malignancy. We propose that AMHR2 may serve as a useful target for vaccination against EOC. To this end, we generated the recombinant 399-amino acid cytoplasmic domain of mouse AMHR2 (AMHR2-CD) and tested its efficacy as a vaccine target in inhibiting growth of the ID8 transplantable EOC cell line in C57BL/6 mice and in preventing growth of autochthonous EOCs that occur spontaneously in transgenic mice. We found that AMHR2-CD immunization of C57BL/6 females induced a prominent antigen-specific proinflammatory CD4+ T cell response that resulted in a mild transient autoimmune oophoritis that resolved rapidly with no detectable lingering adverse effects on ovarian function. AMHR2-CD vaccination significantly inhibited ID8 tumor growth when administered either prophylactically or therapeutically, and protection against EOC growth was passively transferred into naive recipients with AMHR2-CD-primed CD4+ T cells but not with primed B cells. In addition, prophylactic AMHR2-CD vaccination of TgMISIIR-TAg transgenic mice significantly inhibited growth of autochthonous EOCs and provided a 41.7% increase in mean overall survival. We conclude that AMHR2-CD vaccination provides effective immunotherapy of EOC with relatively benign autoimmune complications.
Collapse
|
4
|
Shao J, Zhang J, Wu X, Mao Q, Chen P, Zhu F, Xu M, Kong W, Liang Z, Wang J. Comparing the Primary and Recall Immune Response Induced by a New EV71 Vaccine Using Systems Biology Approaches. PLoS One 2015; 10:e0140515. [PMID: 26465882 PMCID: PMC4605509 DOI: 10.1371/journal.pone.0140515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/25/2015] [Indexed: 12/19/2022] Open
Abstract
Three inactivated EV71 whole-virus vaccines have completed Phase III clinical trials in mainland China, with high efficacy, satisfactory safety, and sustained immunogenicity. However, the molecular mechanisms how this new vaccine elicit potent immune response remain poorly understood. To characterize the primary and recall responses to EV71 vaccines, PBMC from 19 recipients before and after vaccination with EV71 vaccine are collected and their gene expression signatures after stimulation with EV71 antigen were compared. The results showed that primary and recall response to EV71 antigen have both activated an IRF7 regulating type I interferon and antiviral immune response network. However, up-regulated genes involved in T cell activation regulated by IRF1, inflammatory response, B-cell activation and humoral immune response were only observed in recall response. The specific secretion of IL-10 in primary response and IL-2,IP-10,CCL14a, CCL21 in recall response was consistent with the activation of immune response process found in genes. Furthermore, the expression of MX1 and secretion of IP-10 in recall response were strongly correlated with NTAb level at 180d after vaccination (r = 0.81 and 0.99). In summary, inflammatory response, adaptive immune response and a stronger antiviral response were indentified in recall response.
Collapse
Affiliation(s)
- Jie Shao
- School of Life Sciences, Jilin University, Changchun, Jilin, P.R.China
| | - Junnan Zhang
- Division of Hepatitis Virus Vaccines, National Institutes for Food and Drug Control, Beijing, P.R.China
| | - Xing Wu
- Division of Hepatitis Virus Vaccines, National Institutes for Food and Drug Control, Beijing, P.R.China
| | - Qunying Mao
- Division of Hepatitis Virus Vaccines, National Institutes for Food and Drug Control, Beijing, P.R.China
| | - Pan Chen
- Division of Hepatitis Virus Vaccines, National Institutes for Food and Drug Control, Beijing, P.R.China
| | - Fengcai Zhu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R.China
| | - Miao Xu
- Division of Hepatitis Virus Vaccines, National Institutes for Food and Drug Control, Beijing, P.R.China
| | - Wei Kong
- School of Life Sciences, Jilin University, Changchun, Jilin, P.R.China
- * E-mail: (WK); (ZL); (JW)
| | - Zhenglun Liang
- Division of Hepatitis Virus Vaccines, National Institutes for Food and Drug Control, Beijing, P.R.China
- * E-mail: (WK); (ZL); (JW)
| | - Junzhi Wang
- Division of Hepatitis Virus Vaccines, National Institutes for Food and Drug Control, Beijing, P.R.China
- * E-mail: (WK); (ZL); (JW)
| |
Collapse
|
5
|
West NR, McCuaig S, Franchini F, Powrie F. Emerging cytokine networks in colorectal cancer. Nat Rev Immunol 2015; 15:615-29. [PMID: 26358393 DOI: 10.1038/nri3896] [Citation(s) in RCA: 275] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokine networks are crucial aspects of tumour immunology, particularly for colorectal cancer (CRC), in which inflammation and antitumour immunity are key determinants of disease progression. In this Review, we highlight new insights into the functions of well-known cytokines in CRC, describe recently discovered roles for a growing number of novel players, and emphasize the complexity and therapeutic implications of the cytokine milieu. We also discuss how cancer mutations and epigenetic adaptations influence the oncogenic potential of cytokines, a relatively unexplored area that could yield crucial insights into tumour immunology and facilitate the effective application of cytokine-modulatory therapies for CRC.
Collapse
Affiliation(s)
- Nathan R West
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LF, UK.,Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Sarah McCuaig
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LF, UK.,Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Fanny Franchini
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LF, UK.,Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LF, UK.,Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
6
|
Phan GQ, Rosenberg SA. Adoptive cell transfer for patients with metastatic melanoma: the potential and promise of cancer immunotherapy. Cancer Control 2014; 20:289-97. [PMID: 24077405 DOI: 10.1177/107327481302000406] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Current FDA-approved therapeutic options for patients with metastatic melanoma include dacarbazine, interleukin 2, ipilimumab, vemurafenib, dabrafenib, and trametinib, but long-term tumor regression using available agents remains out of reach for most patients. Adoptive cell transfer (ACT) with autologous tumor-infiltrating lymphocytes (TILs) has shown encouraging results in clinical trials, with evidence of durable ongoing complete responses in patients with advanced melanoma. Emerging techniques to engineer T-cell receptors (TCRs) or chimeric antigen receptors (CARs) using lymphocytes from peripheral blood may offer new tactics in ACT. METHODS We reviewed the literature to provide a synopsis on the development and clinical trial results of ACT, as well as the future outlook for using ACT in patients with metastatic melanoma. RESULTS ACT with TILs as part of a lymphodepleting regimen has been shown in clinical trials to cause objective clinical responses in approximately 40% to 72% of patients with metastatic melanoma, with up to 40% of those patients experiencing complete responses lasting up to 7 years ongoing. Pilot trials using TCR-engineered cells against melanoma-associated antigens MART-1 and gp100 and the cancer-testis antigen NY-ESO-1 have shown clinical responses in patients with melanoma. CAR cells directed against melanoma have been tested only in preclinical models; however, CAR cells targeting other histologies such as lymphoma have elicited antitumor responses in patients. CONCLUSIONS An example of state-of-the-art personalized medicine, ACT is a potentially curative therapy for patients with metastatic melanoma. Ongoing trials aiming to simplify the regimens may allow a broader range of patients to be treated and enable ACT to be offered by academic cancer centers.
Collapse
Affiliation(s)
- Giao Q Phan
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
7
|
Coventry BJ, Lilly CA, Hersey P, Michele A, Bright RJ. Prolonged repeated vaccine immuno-chemotherapy induces long-term clinical responses and survival for advanced metastatic melanoma. J Immunother Cancer 2014; 2:9. [PMID: 27437102 PMCID: PMC4950896 DOI: 10.1186/2051-1426-2-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 03/21/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Repetitive long-term Vaccinia Melanoma Cell Lysate (VMCL) vaccination schedules have proved clinically effective in producing Complete Responses and strong durable survivals for up to 6.1 years in a previous study of patients with advanced Stage IV and Stage IIIc melanoma. These studies were expanded to include 54 patients for further evaluation of these findings. METHODS 54 patients comprising 48 Stage IV (6 M1a, 14 M1b, 28 M1c) and 6 advanced Stage III (5 IIIc; 1 IIIb) were studied using repeated intra-dermal VMCL vaccine therapy. If disease progressed, vaccine was continued together with standard chemotherapy (DTIC and/or Fotemustine). Overall survival was the primary end-point assessed, with clinical responses and toxicity recorded. RESULTS From vaccine commencement, median overall survival was 14 months, ranging from 4 to 121 months. Kaplan-Meier survival analysis demonstrated overall 1, 2 and 3-year survival estimates of 57%, 26% and 18.5% respectively, and overall 5-year survival of 15.4%. No appreciable toxicity was observed. Complete Responses (CR) occurred in 16.7% (9) and partial responses (PR) in 14.8% (8) of patients. Stable disease was noted in a further 25 patients (46.3%). No response to therapy was apparent in 12 patients (22.2%). The overall response rate was 31.5% (CR + PR), with clinically significant responses (CR + PR + SD) in 77.8% of patients. Strong, durable clinical responses with overall survivals ≥ 23 months occurred in 29.6% of patients treated with repeated VMCL vaccine for advanced melanoma, (+/- concurrent chemotherapy). CONCLUSIONS Prolonged, repetitive VMCL vaccination immunotherapy appears to be a clinically effective means of generating relatively high CR rates, useful clinical responses and long-term survivals, with little toxicity, but remains notably under-explored. Successive immunomodulation might explain the results. Closer analysis of repetitive dosing is required to improve clinical response rates and survival, perhaps by optimising the timing of immunotherapy delivery. TRIAL REGISTRATION Australian and New Zealand Clinical Trials Registry ANZCTRN12605000425695.
Collapse
Affiliation(s)
- Brendon J Coventry
- Discipline of Surgery, University of Adelaide, Adelaide Melanoma Unit, Royal Adelaide Hospital, Adelaide, South Australia Australia
| | - Carrie A Lilly
- Discipline of Surgery, University of Adelaide, Adelaide Melanoma Unit, Royal Adelaide Hospital, Adelaide, South Australia Australia
| | - Peter Hersey
- Kolling Institute University of Sydney, New South Wales, Australia
| | - Antonio Michele
- Medical Oncology, North Adelaide Oncology, Calvary Hospital, North Adelaide, South Australia 5006 Australia
| | - Richard J Bright
- Discipline of Surgery, University of Adelaide, Adelaide Melanoma Unit, Royal Adelaide Hospital, Adelaide, South Australia Australia
| |
Collapse
|
8
|
Fenoglio D, Traverso P, Parodi A, Kalli F, Zanetti M, Filaci G. Generation of more effective cancer vaccines. Hum Vaccin Immunother 2013; 9:2543-7. [PMID: 23978951 DOI: 10.4161/hv.26147] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cancer vaccines represent a promising therapeutic approach for which prime time is imminent. However, clinical efficacy must be improved in order for cancer vaccines to become a valid alternative or complement to traditional cancer treatments. Considerable efforts have been undertaken so far to better understand the fundamental requirements for clinically-effective cancer vaccines. Recent data emphasize that important requirements, among others, are (1) the use of multi-epitope immunogens, possibly deriving from different tumor antigens; (2) the selection of effective adjuvants; (3) the association of cancer vaccines with agents able to counteract the regulatory milieu present in the tumor microenvironment; and (4) the need to choose the definitive formulation and regimen of a vaccine after accurate preliminary tests comparing different antigen formulations. The first requirement deals with issues related to HLA restriction of tumor antigen presentation, as well as usefulness of tumor antigen spreading and counteraction of immune escape phenomena, linked to tumor antigen down-modulation, for an effective anti-cancer immune response. The second point underscores the necessity of optimal activation of innate immunity to achieve an efficient adaptive anti-cancer immune response. The third point focuses on the importance to inhibit subsets of regulatory cells. The last requirement stresses the concept that the regimen and formulation of the vaccine impacts profoundly on cancer vaccine efficacy. A new generation of cancer vaccines, provided with both immunological and clinical efficacy, will hopefully soon address these requirements.
Collapse
Affiliation(s)
- Daniela Fenoglio
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Internal Medicine; University of Genoa; Genoa, Italy
| | - Paolo Traverso
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Surgical Sciences; University of Genoa; Genoa, Italy
| | - Alessia Parodi
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy
| | - Francesca Kalli
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy
| | - Maurizio Zanetti
- The Laboratory of Immunology; Department of Medicine and Cancer Center; University of California; San Diego, CA USA
| | - Gilberto Filaci
- Centre of Excellence for Biomedical Research; University of Genoa; Genoa, Italy; Department of Internal Medicine; University of Genoa; Genoa, Italy
| |
Collapse
|
9
|
Fenoglio D, Traverso P, Parodi A, Tomasello L, Negrini S, Kalli F, Battaglia F, Ferrera F, Sciallero S, Murdaca G, Setti M, Sobrero A, Boccardo F, Cittadini G, Puppo F, Criscuolo D, Carmignani G, Indiveri F, Filaci G. A multi-peptide, dual-adjuvant telomerase vaccine (GX301) is highly immunogenic in patients with prostate and renal cancer. Cancer Immunol Immunother 2013; 62:1041-1052. [PMID: 23591981 PMCID: PMC11029691 DOI: 10.1007/s00262-013-1415-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 03/07/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Anti-tumor vaccination is a new frontier in cancer treatment applicable to immunogenic neoplasms such as prostate and renal cancers. GX301 is a vaccine constituted by four telomerase peptides and two adjuvants, Montanide ISA-51 and Imiquimod. OBJECTIVE The aim of this study was to analyze safety and tolerability of GX301 in an open-label, phase I/II trial. Immunological and clinical responses were also evaluated as secondary endpoints. EXPERIMENTAL DESIGN GX301 was administered by intradermally injecting 500 μg of each peptide (dissolved in Montanide ISA-51) in the skin of the abdomen. Imiquimod was applied as a cream at the injection sites. The protocol included 8 administrations at days 1, 3, 5, 7, 14, 21, 35, 63. Eligible patients were affected with stage IV prostate or renal cancer resistant to conventional treatments. Patients were clinically and immunologically monitored up to 6 months from the first immunization. RESULTS No grade 3-4 adverse events were observed. Evidence of vaccine-specific immunological responses was detected in 100 % of patients. Disease stabilization occurred in 4 patients. Prolonged progression-free survival and overall survival were observed in patients showing a full pattern of vaccine-specific immunological responses. CONCLUSION GX301 demonstrated to be safe and highly immunogenic. Further studies are needed to determine its clinical efficacy.
Collapse
Affiliation(s)
- Daniela Fenoglio
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Paolo Traverso
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
- Department of Surgical Sciences, University of Genoa, Genoa, Italy
| | - Alessia Parodi
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Laura Tomasello
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Simone Negrini
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Francesca Kalli
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Florinda Battaglia
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Francesca Ferrera
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
| | - Stefania Sciallero
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Maurizio Setti
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Alberto Sobrero
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Francesco Boccardo
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Giuseppe Cittadini
- Istituto Nazionale per la Ricerca sul Cancro, IRCCS Azienda Ospedaliero Universitaria San Martino—IST, Genoa, Italy
| | - Francesco Puppo
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Domenico Criscuolo
- Genovax srl, Colleretto Giacosa, Italy
- Present Address: Mediolanum Farmaceutici Spa, Milan, Italy
| | | | - Francesco Indiveri
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Gilberto Filaci
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV n. 7, 16132 Genoa, Italy
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
10
|
Kalli F, Machiorlatti R, Battaglia F, Parodi A, Conteduca G, Ferrera F, Proietti M, Tardito S, Sanguineti M, Millo E, Fenoglio D, De Palma R, Inghirami G, Filaci G. Comparative analysis of cancer vaccine settings for the selection of an effective protocol in mice. J Transl Med 2013; 11:120. [PMID: 23663506 PMCID: PMC3659084 DOI: 10.1186/1479-5876-11-120] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/08/2013] [Indexed: 12/31/2022] Open
Abstract
Background Cancer vaccines are considered a promising therapeutic approach. However, their clinical results are not yet satisfactory. This may be due to the the difficulty of selection of an efficient tumor associated antigen (TAA) and immunization protocol. Indeed, the weak antigenicity of many TAA impairs the design of robust procedures, therefore a systematic analysis to identify the most efficient TAA is mandatory. Here, we performed a study to compare different gp100 vaccination strategies to identify the best strategy to provide a 100% protection against experimental melanoma in a reproducible manner. Methods C57BL/6J mice were challenged subcutaneously with B16F10 melanoma cells, after vaccination with: a) mouse or human gp10025-33 peptide plus CpG adjuvant; b) mouse or human gp100 gene; c) mouse or human gp10025-33 peptide-pulsed dendritic cells (DC). Alternatively, a neutralizing anti-IL-10 monoclonal antibody (mAb) was subcutaneously administered at the site of tumor challenge to counteract regulatory cells. Finally, combinatorial treatment was performed associating human gp10025-33 peptide-pulsed DC vaccination with administration of the anti-IL-10 mAb. Results Vaccination with human gp10025-33 peptide-pulsed DC was the most effective immunization protocol, although not achieving a full protection. Administration of the anti-IL-10 mAb showed also a remarkable protective effect, replicated in mice challenged with a different tumor, Anaplastic Large Cell Lymphoma. When immunization with gp10025-33 peptide-pulsed DC was associated with IL-10 counteraction, a 100% protective effect was consistently achieved. The analysis on the T-cell tumor infiltrates showed an increase of CD4+granzyme+ T-cells and a decreased number of CD4+CD25+Foxp3+ Treg elements from mice treated with either gp10025-33 peptide-pulsed DC vaccination or anti-IL-10 mAb administration. These data suggest that processes of intratumoral re-balance between effector and regulatory T cell subpopulations may play a critical protective role in immunotherapy protocols. Conclusions Here we demonstrate that, in the setting of a cancer vaccine strategy, a comparative analysis of different personalized approaches may favour the unveiling of the most effective protocol. Moreover, our findings suggest that counteraction of IL-10 activity may be critical to revert the intratumoral environment promoting Treg polarization, thus increasing the effects of a vaccination against selected TAA.
Collapse
Affiliation(s)
- Francesca Kalli
- Centre of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV n. 7, 16132, Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Filipazzi P, Pilla L, Mariani L, Patuzzo R, Castelli C, Camisaschi C, Maurichi A, Cova A, Rigamonti G, Giardino F, Di Florio A, Asioli M, Frati P, Sovena G, Squarcina P, Maio M, Danielli R, Chiarion-Sileni V, Villa A, Lombardo C, Tragni G, Santinami M, Parmiani G, Rivoltini L. Limited induction of tumor cross-reactive T cells without a measurable clinical benefit in early melanoma patients vaccinated with human leukocyte antigen class I-modified peptides. Clin Cancer Res 2012; 18:6485-96. [PMID: 23032742 DOI: 10.1158/1078-0432.ccr-12-1516] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The progressive immune dysfunctions that occur in patients with advanced melanoma make them unlikely to efficiently respond to cancer vaccines. A multicenter randomized phase II trial was conducted to test whether immunization with modified HLA class I tumor peptides in the context of adjuvant therapy results in better immunologic responses and improved clinical outcomes in patients with early melanoma (stages IIB/C-III). EXPERIMENTAL DESIGN Forty-three patients were enrolled to undergo vaccination (n = 22) or observation (n = 21). The vaccine included four HLA-A*0201-restricted modified peptides (Melan-A/MART-1([27L]), gp100([210M]), NY-ESO-1([165V]), and Survivin([97M])) emulsified in Montanide ISA51 and injected subcutaneously in combination with cyclophosphamide (300 mg/m(2)) and low-dose IL-2 (3 × 10(6) IU). The immune responses were monitored using ex vivo IFN-γ-ELISpot, HLA/multimer staining, and in vitro short-term peptide sensitization assays. RESULTS Vaccination induced a rapid and persistent increase in specific effector memory CD8(+) T cells in 75% of the patients. However, this immunization was not associated with any significant increase in disease-free or overall survival as compared with the observation group. An extensive immunologic analysis revealed a significantly reduced cross-recognition of the corresponding native peptides and, most importantly, a limited ability to react to melanoma cells. CONCLUSIONS Adjuvant setting is an appealing approach for testing cancer vaccines because specific CD8(+) T cells can be efficiently induced in most vaccinated patients. However, the marginal antitumor activity of the T cells induced by modified peptides in this study largely accounts for the observed lack of benefit of vaccination. These findings suggest reconsidering this immunization strategy, particularly in early disease.
Collapse
Affiliation(s)
- Paola Filipazzi
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|