1
|
Isaak AJ, Clements GR, Buenaventura RGM, Merlino G, Yu Y. Development of Personalized Strategies for Precisely Battling Malignant Melanoma. Int J Mol Sci 2024; 25:5023. [PMID: 38732242 PMCID: PMC11084485 DOI: 10.3390/ijms25095023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Melanoma is the most severe and fatal form of skin cancer, resulting from multiple gene mutations with high intra-tumor and inter-tumor molecular heterogeneity. Treatment options for patients whose disease has progressed beyond the ability for surgical resection rely on currently accepted standard therapies, notably immune checkpoint inhibitors and targeted therapies. Acquired resistance to these therapies and treatment-associated toxicity necessitate exploring novel strategies, especially those that can be personalized for specific patients and/or populations. Here, we review the current landscape and progress of standard therapies and explore what personalized oncology techniques may entail in the scope of melanoma. Our purpose is to provide an up-to-date summary of the tools at our disposal that work to circumvent the common barriers faced when battling melanoma.
Collapse
Affiliation(s)
| | | | | | | | - Yanlin Yu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Li C, Wang S, Ma X, Wang T, Lu R, Jia X, Leng Z, Kong X, Zhang J, Li L. Ranitidine as an adjuvant regulates macrophage polarization and activates CTLs through the PI3K-Akt2 signaling pathway. Int Immunopharmacol 2023; 116:109729. [PMID: 37800555 DOI: 10.1016/j.intimp.2023.109729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 02/19/2023]
Abstract
Adjuvants are an indispensable component of vaccines, but there are few adjuvants for human vaccines. H2 receptor blockers, inhibiting gastric acid secretion, have immune enhancement effects. Ranitidine (RAN) is a water-soluble H2 receptor blocker, and whether it has an immune-enhancing effect is still unknown. In this study, flow cytometry, western blotting, and immunofluorescence methods were used to analyze whether RAN could activate macrophage polarization to the M1 phenotype in vivo and in vitro. Here, we found that the M1 inflammatory cytokine levels and surface markers in RAW264.7 cells were upregulated by NF-κB activation, possibly through the PI3K-Akt2 signaling pathway, after RAN treatment. Endocytic function was also enhanced by feedback regulation of Akt2/GSK3β/Dynmin1 signaling. Furthermore, to evaluate the adjuvant function of RAN, we used OVA plus RAN as a vaccine to inhibit the growth of B16-OVA tumors in mice. We also found that in the RAN adjuvant group, macrophage polarization to M1, Th1 cell differentiation, and cytotoxic T lymphocyte (CTL) activation were significantly upregulated. The tumor growth of mice was inhibited, and the survival rate of mice was significantly improved. This study provides new evidence for the mechanism by which RAN activates the immune response and is expected to provide a new strategy for the research and development of tumor vaccine adjuvants.
Collapse
Affiliation(s)
- Chenglin Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Shuang Wang
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China.
| | - Xiaoran Ma
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Tiantian Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Ran Lu
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Xihui Jia
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Zhe Leng
- Department of Gynecology, Qingdao Women and Children's Hospital, Qingdao 266000, China
| | - Xiaowen Kong
- School of Stomatology, Qingdao University, Qingdao 266071, China
| | - Jinyu Zhang
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China
| | - Ling Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong, China.
| |
Collapse
|
3
|
Jazowiecka-Rakus J, Sochanik A, Rusin A, Hadryś A, Fidyk W, Villa N, Rahman MM, Chmielik E, Franco LS, McFadden G. Myxoma Virus-Loaded Mesenchymal Stem Cells in Experimental Oncolytic Therapy of Murine Pulmonary Melanoma. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:335-350. [PMID: 32775618 PMCID: PMC7398944 DOI: 10.1016/j.omto.2020.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Oncolytic viruses can target neoplasms, triggering oncolytic and immune effects. Their delivery to melanoma lesions remains challenging. Bone-marrow-derived mesenchymal stem cells (MSCs) were shown to be permissive for oncolytic myxoma virus (MYXV), allowing its transfer to melanoma cells, leading to their killing. Involvement of progeny virus was demonstrated in the transfer from MSCs to co-cultured melanoma cells. The inhibitory effect of virus on melanoma foci formation in murine lungs was revealed using melanoma cells previously co-cultured with MYXV-infected MSCs. Virus accumulation and persistence in lungs of lesion-bearing mice were shown following intravenous administration of MSC-shielded MYXV construct encoding luciferase. Therapy of experimentally induced lung melanoma in mice with interleukin (IL)-15-carrying MYXV construct delivered by MSCs led to marked regression of lesions and could increase survival. Elevated natural killer (NK) cell percentages in blood indicated robust innate responses against unshielded virus only. Lung infiltration by NK cells was followed by inflow of CD8+ T lymphocytes into melanoma lesions. Elevated expression of genes involved in adaptive immune response following oncolytic treatment was confirmed using RT-qPCR. No adverse pathological effects related to MSC-mediated oncolytic therapy with MYXV were observed. MSCs allow for safe and efficient ferrying of therapeutic MYXV to pulmonary melanoma foci triggering immune effects.
Collapse
Affiliation(s)
- Joanna Jazowiecka-Rakus
- Maria Skłodowska-Curie Memorial National Research Institute of Oncology, 44-102 Gliwice, Poland
- Corresponding author: Joanna Jazowiecka-Rakus, Maria Skłodowska-Curie Memorial National Research Institute of Oncology, 44-102 Gliwice, Poland.
| | - Aleksander Sochanik
- Maria Skłodowska-Curie Memorial National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Aleksandra Rusin
- Maria Skłodowska-Curie Memorial National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Agata Hadryś
- Maria Skłodowska-Curie Memorial National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Wojciech Fidyk
- Maria Skłodowska-Curie Memorial National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Nancy Villa
- Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | | | - Ewa Chmielik
- Maria Skłodowska-Curie Memorial National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Lina S. Franco
- Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Grant McFadden
- Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
4
|
Podaza E, Carri I, Aris M, von Euw E, Bravo AI, Blanco P, Ortiz Wilczyñski JM, Koile D, Yankilevich P, Nielsen M, Mordoh J, Barrio MM. Evaluation of T-Cell Responses Against Shared Melanoma Associated Antigens and Predicted Neoantigens in Cutaneous Melanoma Patients Treated With the CSF-470 Allogeneic Cell Vaccine Plus BCG and GM-CSF. Front Immunol 2020; 11:1147. [PMID: 32582212 PMCID: PMC7290006 DOI: 10.3389/fimmu.2020.01147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/11/2020] [Indexed: 01/30/2023] Open
Abstract
The CSF-470 vaccine consists of lethally-irradiated allogeneic cells derived from four cutaneous melanoma cell lines administered plus BCG and GM-CSF as adjuvants. In an adjuvant phase II study vs. IFN-α2b, the vaccine significantly prolonged the distant metastasis-free survival (DMFS) of stages IIB-IIC-III melanoma patients with evidence of the induction of immune responses against vaccine cells. Purpose: The aim of this study was to analyze the antigens against which the immune response was induced, as well as the T-helper profile and lytic ability of immune cells after CSF-470 treatment. Methods: HLA-restricted peptides from tumor-associated antigens (TAAs) were selected from TANTIGEN database for 13 evaluable vaccinated patients. In addition, for patient #006 (pt#006), tumor somatic variants were identified by NGS and candidate neoAgs were selected by predicted HLA binding affinity and similarity between wild type (wt) and mutant peptides. The patient's PBMC reactivity against selected peptides was detected by IFNγ-ELISPOT. T-helper transcriptional profile was determined by quantifying GATA-3, T-bet, and FOXP3 mRNA by RT-PCR, and intracellular cytokines were analyzed by flow cytometry. Autologous tumor cell lysis by PBMC was assessed in an in vitro calcein release assay. Results: Vaccinated patient's PBMC reactivity against selected TAAs derived peptides showed a progressive increase in the number of IFNγ-producing cells throughout the 2-yr vaccination protocol. ELISPOT response correlated with delayed type hypersensitivity (DTH) reaction to CSF-470 vaccine cells. Early upregulation of GATA-3 and Foxp3 mRNA, as well as an increase in CD4+IL4+cells, was associated with a low DMFS. Also, IFNγ response against 9/73 predicted neoAgs was evidenced in the case of pt#006; 7/9 emerged after vaccination. We verified in pt# 006 that post-vaccination PBMC boosted in vitro with the vaccine lysate were able to lyse autologous tumor cells. Conclusions: A progressive increase in the immune response against TAAs expressed in the vaccine and in the patient's tumor was induced by CSF-470 vaccination. In pt#006, we demonstrated immune recognition of patient's specific neoAgs, which emerged after vaccination. These results suggest that an initial response against shared TAAs could further stimulate an immune response against autologous tumor neoAgs.
Collapse
Affiliation(s)
- Enrique Podaza
- Centro de Investigaciones Oncológicas, Fundación Cáncer, Buenos Aires, Argentina
| | - Ibel Carri
- IIBIO, UNSAM, San Martín, Buenos Aires, Argentina
| | - Mariana Aris
- Centro de Investigaciones Oncológicas, Fundación Cáncer, Buenos Aires, Argentina
| | - Erika von Euw
- UCLA JCCC-Translational Oncology Research Labs, Los Angeles, CA, United States
- T-Cure Bioscience Inc., Los Angeles, CA, United States
| | - Alicia Inés Bravo
- Centro de Excelencia en Medicina Translacional, Hospital El Cruce, Buenos Aires, Argentina
| | - Paula Blanco
- Centro de Investigaciones Oncológicas, Fundación Cáncer, Buenos Aires, Argentina
| | | | - Daniel Koile
- Plataforma Bioinformática, INBioBA-MPSP, Buenos Aires, Argentina
| | | | - Morten Nielsen
- IIBIO, UNSAM, San Martín, Buenos Aires, Argentina
- Department of Health Technology, The Technical University of Denmark, Lyngby, Denmark
| | - José Mordoh
- Centro de Investigaciones Oncológicas, Fundación Cáncer, Buenos Aires, Argentina
- IIBBA-CONICET, Fundación Instituto Leloir, Buenos Aires, Argentina
- Instituto Alexander Fleming, Buenos Aires, Argentina
| | - María Marcela Barrio
- Centro de Investigaciones Oncológicas, Fundación Cáncer, Buenos Aires, Argentina
| |
Collapse
|
5
|
Benitez MLR, Bender CB, Oliveira TL, Schachtschneider KM, Collares T, Seixas FK. Mycobacterium bovis BCG in metastatic melanoma therapy. Appl Microbiol Biotechnol 2019; 103:7903-7916. [PMID: 31402426 DOI: 10.1007/s00253-019-10057-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
Melanoma is the most aggressive form of skin cancer, with a high mortality rate and with 96,480 new cases expected in 2019 in the USS. BRAFV600E, the most common driver mutation, is found in around 50% of melanomas, contributing to tumor growth, angiogenesis, and metastatic progression. Dacarbazine (DTIC), an alkylate agent, was the first chemotherapeutic agent approved by the US Food and Drug Administration (FDA) used as a standard treatment. Since then, immunotherapies have been approved for metastatic melanoma (MM) including ipilimumab and pembrolizumab checkpoint inhibitors that help decrease the risk of progression. Moreover, Mycobacterium bovis Bacillus Calmette-Guerin (BCG) serves as an adjuvant therapy that induces the recruitment of natural killer NK, CD4+, and CD8+ T cells and contributes to antitumor immunity. BCG can be administered in combination with chemotherapeutic and immunotherapeutic agents and can be genetically manipulated to produce recombinant BCG (rBCG) strains that express heterologous proteins or overexpress immunogenic proteins, increasing the immune response and improving patient survival. In this review, we highlight several studies utilizing rBCG immunotherapy for MM in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Martha Lucia Ruiz Benitez
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Camila Bonnemann Bender
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Thaís Larré Oliveira
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Tiago Collares
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabiana Kömmling Seixas
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
6
|
Pampena MB, Barrio MM, Juliá EP, Blanco PA, von Euw EM, Mordoh J, Levy EM. Early Events of the Reaction Elicited by CSF-470 Melanoma Vaccine Plus Adjuvants: An In Vitro Analysis of Immune Recruitment and Cytokine Release. Front Immunol 2017; 8:1342. [PMID: 29109725 PMCID: PMC5660290 DOI: 10.3389/fimmu.2017.01342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/03/2017] [Indexed: 11/13/2022] Open
Abstract
In a previous work, we showed that CSF-470 vaccine plus bacillus Calmette–Guerin (BCG) and granulocyte macrophage colony-stimulating factor (GM-CSF) as adjuvants resulted in a significant benefit in the distant metastasis-free survival when comparing vaccinated vs. IFN-α2b-treated high-risk cutaneous melanoma patients in a Phase II study. Immune monitoring demonstrated an increase in anti-tumor innate and adaptive immunities of vaccinated patients, with a striking increase in IFN-γ secreting lymphocytes specific for melanoma antigens (Ags). In an effort to dissect the first steps of the immune response elicited by CSF-470 vaccine plus adjuvants, we evaluated, in an in vitro model, leukocyte migration, cytokine production, and monocyte phagocytosis of vaccine cells. Our results demonstrate that leukocytes recruitment, mostly from the innate immune system, is an early event after CSF-470 vaccine plus BCG plus GM-CSF interaction with immune cells, possibly explained by the high expression of CCL2/MCP-1 and other chemokines by vaccine cells. Early release of TNF-α and IL-1β pro-inflammatory cytokines and efficient tumor Ags phagocytosis by monocytes take place and would probably create a favorable context for Ag processing and presentation. Although the presence of the vaccine cells hampered cytokines production stimulated by BCG in a mechanism partially mediated by TGF-β and IL-10, still significant levels of TNF-α and IL-1β could be detected. Thus, BCG was required to induce local inflammation in the presence of CSF-470 vaccine cells.
Collapse
Affiliation(s)
- María B Pampena
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - María M Barrio
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - Estefanía P Juliá
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - Paula A Blanco
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - Erika M von Euw
- UCLA JCCC-Translational Oncology Research Labs, Los Angeles, CA, United States
| | - José Mordoh
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina.,Instituto Médico Especializado Alexander Fleming, Buenos Aires, Argentina.,Fundación Instituto Leloir, IIBBA-CONICET, Buenos Aires, Argentina
| | | |
Collapse
|
7
|
Bräunlein E, Krackhardt AM. Tools to define the melanoma-associated immunopeptidome. Immunology 2017; 152:536-544. [PMID: 28755382 DOI: 10.1111/imm.12803] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 07/25/2017] [Accepted: 07/25/2017] [Indexed: 12/26/2022] Open
Abstract
Immunotherapies have been traditionally applied in malignant melanoma, which represent one of the most immunogenic tumours. Recently, immune checkpoint modulation has shown high therapeutic efficacy and may provide long-term survival in a significant proportion of affected patients. T cells are the major players in tumour rejection and recognize tumour cells predominantly in an MHC-dependent way. The immunopeptidome comprises the peptide repertoire presented by MHC class I and II molecules on the surface of the body's cells including tumour cells. To understand characteristics of suitable rejection antigens as well as respective effective T-cell responses, determination of the immunopeptidome is of utmost importance. Suitable rejection antigens need to be further characterized and validated not only to systematically improve current therapeutic approaches, but also to develop individualized treatment options. In this review, we report on current tools to explore the immunopeptidome in human melanoma and discuss current understanding and future developments to specifically detect and select those antigens that may be most relevant and promising for effective tumour rejection.
Collapse
Affiliation(s)
- Eva Bräunlein
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Angela M Krackhardt
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,German Cancer Consortium of Translational Cancer Research (DKTK) and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
8
|
Mordoh J, Pampena MB, Aris M, Blanco PA, Lombardo M, von Euw EM, Mac Keon S, Yépez Crow M, Bravo AI, O'Connor JM, Orlando AG, Ramello F, Levy EM, Barrio MM. Phase II Study of Adjuvant Immunotherapy with the CSF-470 Vaccine Plus Bacillus Calmette-Guerin Plus Recombinant Human Granulocyte Macrophage-Colony Stimulating Factor vs Medium-Dose Interferon Alpha 2B in Stages IIB, IIC, and III Cutaneous Melanoma Patients: A Single Institution, Randomized Study. Front Immunol 2017; 8:625. [PMID: 28620382 PMCID: PMC5449770 DOI: 10.3389/fimmu.2017.00625] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 05/10/2017] [Indexed: 12/27/2022] Open
Abstract
The irradiated, allogeneic, cellular CSF-470 vaccine plus Bacillus Calmette–Guerin (BCG) and recombinant human granulocyte macrophage-colony stimulating factor (rhGM-CSF) is being tested against medium-dose IFN-α2b in stages IIB–III cutaneous melanoma (CM) patients (pts) after surgery in an open, randomized, Phase II/III study. We present the results of the Phase II part of the ongoing CASVAC-0401 study (ClinicalTrials.gov: NCT01729663). Thirty-one pts were randomized to the CSF-470 vaccine (n = 20) or to the IFN-α2b arm (n = 11). During the 2-year treatment, immunized pts should receive 13 vaccinations. On day 1 of each visit, 1.6 × 107 irradiated CSF-470 cells plus 106 colony-forming units BCG plus 100 µg rhGM-CSF were administered intradermally, followed on days 2–4 by 100 µg rhGM-CSF. IFN-α2b pts should receive 10 million units (MU)/day/5 days a week for 4 weeks; then 5 MU thrice weekly for 23 months. Toxicity and quality of life (QOL) were evaluated at each visit. With a mean and a maximum follow-up of 39.4 and 83 months, respectively, a significant benefit in the distant metastasis-free survival (DMFS) for CSF-470 was observed (p = 0.022). Immune monitoring showed an increase in antitumoral cellular and humoral response in vaccinated pts. CSF-470 was well tolerated; 20/20 pts presented grades 1–2 dermic reactions at the vaccination site; 3/20 pts presented grade 3 allergic reactions. Other adverse events (AEs) were grade 1. Pts in the IFN-α2b arm presented grades 2–3 hematological (7/11), hepatic (2/11), and cardiac (1/11) toxicity; AEs in 9/11 pts forced treatment interruptions. QOL was significantly superior in the vaccine arm (p < 0.0001). Our results suggest that CSF-470 vaccine plus BCG plus GM-CSF can significantly prolong, with lower toxicity, the DMFS of high-risk CM pts with respect to medium-dose IFN-α2b. The continuation of a Phase III part of the CASVAC-0401 study is encouraged.
Collapse
Affiliation(s)
- José Mordoh
- Instituto Médico Especializado Alexander Fleming, Buenos Aires, Argentina.,Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina.,Fundación Instituto Leloir, IIBBA-CONICET, Buenos Aires, Argentina
| | | | - Mariana Aris
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | | | | | - Erika María von Euw
- UCLA JCCC-Translational Oncology Research Labs, Los Angeles, CA, United States
| | - Soledad Mac Keon
- Fundación Instituto Leloir, IIBBA-CONICET, Buenos Aires, Argentina
| | - Michelle Yépez Crow
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - Alicia Inés Bravo
- Unidad de Inmunopatología, Hospital Interzonal General de Agudos Eva Perón, San Martín, Argentina
| | | | - Ana Gabriela Orlando
- Hospital Central Olga Rizzi, Reconquista, Argentina.,Instituto Oncológico Mater Dei, Reconquista, Argentina
| | | | | | | |
Collapse
|
9
|
Metastatic melanoma treatment: Combining old and new therapies. Crit Rev Oncol Hematol 2015; 98:242-53. [PMID: 26616525 DOI: 10.1016/j.critrevonc.2015.11.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 10/16/2015] [Accepted: 11/12/2015] [Indexed: 01/04/2023] Open
Abstract
Metastatic melanoma is an aggressive form of cancer characterised by poor prognosis and a complex etiology. Until 2010, the treatment options for metastatic melanoma were very limited. Largely ineffective dacarbazine, temozolamide or fotemustine were the only agents in use for 35 years. In recent years, the development of molecularly targeted inhibitors in parallel with the development of checkpoint inhibition immunotherapies has rapidly improved the outcomes for metastatic melanoma patients. Despite these new therapies showing initial promise; resistance and poor duration of response have limited their effectiveness as monotherapies. Here we provide an overview of the history of melanoma treatment, as well as the current treatments in development. We also discuss the future of melanoma treatment as we go beyond monotherapies to a combinatorial approach. Combining older therapies with the new molecular and immunotherapies will be the most promising way forward for treatment of metastatic melanoma.
Collapse
|
10
|
Gupta S, Termini JM, Rivas Y, Otero M, Raffa FN, Bhat V, Farooq A, Stone GW. A multi-trimeric fusion of CD40L and gp100 tumor antigen activates dendritic cells and enhances survival in a B16-F10 melanoma DNA vaccine model. Vaccine 2015; 33:4798-806. [PMID: 26241951 DOI: 10.1016/j.vaccine.2015.07.081] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/21/2015] [Accepted: 07/25/2015] [Indexed: 12/21/2022]
Abstract
Vaccination with tumor-associated antigens can induce cancer-specific CD8+ T cells. A recent improvement has been the targeting of antigen to dendritic cells (DC) using antibodies that bind DC surface molecules. This study explored the use of multi-trimers of CD40L to target the gp100 melanoma tumor antigen to DC. The spontaneously-multimerizing gene Surfactant Protein D (SPD) was used to fuse gp100 tumor antigen and CD40L, creating the recombinant protein SPD-gp100-CD40L. This "third generation" DC-targeting vaccine was designed to both target antigen to DC and optimally activate dendritic cells by aggregating CD40 trimers on the DC membrane surface. SPD-gp100-CD40L expressed as a 110kDa protein. Analytical light scattering analysis gave elution data corresponding to 4-trimer and multi-trimer SPD-gp100-CD40L oligomers. The protein was biologically active on dendritic cells and induced CD40-mediated NF-κB signaling. DNA vaccination with SPD-gp100-CD40L plasmid, together with plasmids encoding IL-12p70 and GM-CSF, significantly enhanced survival and inhibited tumor growth in a B16-F10 melanoma model. Expression of gp100 and SPD-CD40L as separate molecules did not enhance survival, highlighting the requirement to encode gp100 within SPD-CD40L for optimal vaccine activity. These data support a model where DNA vaccination with SPD-gp100-CD40L targets gp100 to DC in situ, induces activation of these DC, and generates a protective anti-tumor response when given in combination with IL-12p70 and GM-CSF plasmids.
Collapse
Affiliation(s)
- Sachin Gupta
- Department of Microbiology and Immunology, Miami Center for AIDS Research, and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - James M Termini
- Department of Microbiology and Immunology, Miami Center for AIDS Research, and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Yaelis Rivas
- Department of Microbiology and Immunology, Miami Center for AIDS Research, and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Miguel Otero
- Department of Microbiology and Medical Zoology, University of Puerto Rico, San Juan, PR, USA
| | - Francesca N Raffa
- Department of Microbiology and Immunology, Miami Center for AIDS Research, and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vikas Bhat
- Department of Biochemistry and Molecular Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amjad Farooq
- Department of Biochemistry and Molecular Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Geoffrey W Stone
- Department of Microbiology and Immunology, Miami Center for AIDS Research, and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
11
|
Jarosz-Biej M, Smolarczyk R, Cichoń T, Kułach N, Czapla J, Matuszczak S, Szala S. Combined Tumor Cell-Based Vaccination and Interleukin-12 Gene Therapy Polarizes the Tumor Microenvironment in Mice. Arch Immunol Ther Exp (Warsz) 2015; 63:451-64. [PMID: 25801067 PMCID: PMC4633448 DOI: 10.1007/s00005-015-0337-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/09/2015] [Indexed: 12/22/2022]
Abstract
Tumor progression depends on tumor milieu, which influences neovasculature formation and immunosuppression. Combining immunotherapy with antiangiogenic/antivascular therapy might be an effective therapeutic approach. The aim of our study was to elaborate an anticancer therapeutic strategy based on the induction of immune response which leads to polarization of tumor milieu. To achieve this, we developed a tumor cell-based vaccine. CAMEL peptide was used as a B16-F10 cell death-inducing agent. The lysates were used as a vaccine to immunize mice bearing B16-F10 melanoma tumors. To further improve the therapeutic effect of the vaccine, we combined it with interleukin (IL)-12 gene therapy. IL-12, a cytokine with antiangiogenic properties, activates nonspecific and specific immune responses. We observed that combined therapy is significantly more effective (as compared with monotherapies) in inhibiting tumor growth. Furthermore, the tested combination polarizes the tumor microenvironment, which results in a switch from a proangiogenic/immunosuppressive to an antiangiogenic/immunostimulatory one. The switch manifests itself as a decreased number of tumor blood vessels, increased levels of tumor-infiltrating CD4+, CD8+ and NK cells, as well as lower level of suppressor lymphocytes (Treg). Our results suggest that polarizing tumor milieu by such combined therapy does inhibit tumor growth and seems to be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland.
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Natalia Kułach
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
- Department of Animal Physiology and Ecotoxycology, University of Silesia, Katowice, Poland
| | - Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Sybilla Matuszczak
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Stanisław Szala
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| |
Collapse
|
12
|
Tosti G, Cocorocchio E, Pennacchioli E, Ferrucci PF, Testori A, Martinoli C. Heat-shock proteins-based immunotherapy for advanced melanoma in the era of target therapies and immunomodulating agents. Expert Opin Biol Ther 2014; 14:955-67. [PMID: 24670226 DOI: 10.1517/14712598.2014.902928] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Heat-shock proteins (HSPs) are highly conserved, stress-induced proteins that function as chaperones, stabilizing proteins and delivering peptides. Tumor-derived HSP peptide complexes (HSPPCs) induced immunity against several malignancies in preclinical models, exhibiting activity across tumor types. AREAS COVERED HSPPC-based vaccination showed clinical activity in subsets of patients with different malignancies (e.g., gastric, colorectal, pancreatic, ovarian cancer, and glioblastoma). In Phase III clinical trials for advanced melanoma and renal cell carcinoma patients, HSPPC-based vaccine demonstrated an excellent safety profile, thus emerging as a flexible tumor- and patient-specific therapeutic approach. EXPERT OPINION Melanoma, renal clear cell carcinoma, and glioblastoma are among suitable targets for HSP-based treatment as demonstrated by immune responses and clinical activity observed in subsets of patients, mainly those with early stage of disease and limited tumor burden. In order to further improve clinical activity, combinations of HSPPC-based vaccines with mutation-driven therapies, antiangiogenic agents, or immunomodulating monoclonal antibodies should be tested in controlled clinical trials.
Collapse
Affiliation(s)
- Giulio Tosti
- Istituto Europeo di Oncologia, Melanoma and Sarcoma Division , Via Ripamonti 435, 2014i Milano , Italy +39 02 57489459 ; +39 02 94379230 ;
| | | | | | | | | | | |
Collapse
|
13
|
Tomei S, Wang E, Delogu LG, Marincola FM, Bedognetti D. Non-BRAF-targeted therapy, immunotherapy, and combination therapy for melanoma. Expert Opin Biol Ther 2014; 14:663-86. [DOI: 10.1517/14712598.2014.890586] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
14
|
Kaufman KL, Mactier S, Armstrong NJ, Mallawaaratchy D, Byrne SN, Haydu LE, Jakrot V, Thompson JF, Mann GJ, Scolyer RA, Christopherson RI. Surface antigen profiles of leukocytes and melanoma cells in lymph node metastases are associated with survival in AJCC stage III melanoma patients. Clin Exp Metastasis 2014; 31:407-21. [PMID: 24435119 PMCID: PMC3973954 DOI: 10.1007/s10585-014-9636-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 01/09/2014] [Indexed: 12/14/2022]
Abstract
There is an urgent need to identify more accurate prognostic biomarkers in melanoma patients, particularly in those with metastatic disease. This study aimed to identify melanoma and leukocyte surface antigens predictive of survival in a prospective series of AJCC stage IIIb/c melanoma patients (n = 29). Live cell suspensions were prepared from melanoma metastases within lymph nodes (LN). The suspensions were immuno-magnetically separated into CD45+ (leukocyte) and CD45− (non-hematopoietic, enriched melanoma cell) fractions. Surface antigens on CD45− and CD45+ cell populations were profiled using DotScan™ microarrays (Medsaic Pty. Ltd.) and showed differential abundance levels for 52 and 78 antigens respectively. Associations of the surface profiles with clinicopathologic and outcome data (median follow-up 35.4 months post LN resection) were sought using univariate (log-rank test) and multivariate (Wald’s test; modelled with patient’s age, gender and AJCC staging at LN recurrence) survival models. CD9 (p = 0.036), CD39 (p = 0.004) and CD55 (p = 0.005) on CD45+ leukocytes were independently associated with distant metastasis-free survival using multivariate analysis. Leukocytes with high CD39 levels were also significantly associated with increased overall survival (OS) in multivariate analysis (p = 0.016). LNs containing leukocytes expressing CD11b (p = 0.025), CD49d (p = 0.043) and CD79b (p = 0.044) were associated with reduced OS on univariate analysis. For enriched melanoma cells (CD45− cell populations), 11 surface antigens were significantly correlated with the disease-free interval (DFI) between diagnosis of culprit primary melanoma and LN metastasis resection. Nine antigens on CD45+ leukocytes also correlated with DFI. Following validation in independent datasets, surface markers identified here should enable more accurate determination of prognosis in stage III melanoma patients and provide better risk stratification of patients entering clinical trials.
Collapse
Affiliation(s)
- Kimberley L Kaufman
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Swetlana Mactier
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia
| | - Nicola J Armstrong
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,School of Mathematics and Statistics and Prince of Wales Clinical School, University of New South Wales, Kensington, NSW, 2052, Australia
| | | | - Scott N Byrne
- Discipline of Infectious Diseases and Immunology Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia.,Discipline of Dermatology, Bosch Institute, Faculty of Medicine, Sydney Medical School, Sydney, NSW, 2006, Australia
| | - Lauren E Haydu
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Discipline of Surgery, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Valerie Jakrot
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia
| | - John F Thompson
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia
| | - Graham J Mann
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Westmead Institute of Cancer Research, The University of Sydney at Westmead Millennium Institute, Westmead, NSW, 2145, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia.,Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | | |
Collapse
|
15
|
Nicolay HJM, Sigalotti L, Fonsatti E, Covre A, Parisi G, Fratta E, Coral S, Maio M. Epigenetically regulated tumor-associated antigens in melanoma. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/edm.09.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
16
|
Möller MG, Salwa S, Soden DM, O’Sullivan GC. Electrochemotherapy as an adjunct or alternative to other treatments for unresectable or in-transit melanoma. Expert Rev Anticancer Ther 2014; 9:1611-30. [DOI: 10.1586/era.09.129] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
|
18
|
Miguel A, Herrero MJ, Sendra L, Botella R, Algás R, Sánchez M, Aliño SF. Comparative antitumor effect among GM-CSF, IL-12 and GM-CSF+IL-12 genetically modified tumor cell vaccines. Cancer Gene Ther 2013; 20:576-81. [PMID: 23969885 DOI: 10.1038/cgt.2013.54] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 07/03/2013] [Indexed: 02/06/2023]
Abstract
Genetically modified cells have been shown to be one of the most effective cancer vaccine strategies. An evaluation is made of the efficacy of both preventive and therapeutic antitumor vaccines against murine melanoma, using C57BL/6 mice and irradiated B16 tumor cells expressing granulocyte and macrophage colony-stimulating factor (GM-CSF), interleukin-12 (IL-12) or both. Tumor was transplanted by the injection of wild-type B16 cells. Tumor growth and survival were measured to evaluate the efficacy of vaccination. Specific humoral response and immunoglobulin G (IgG) switch were evaluated measuring total IgG and IgG1 and IgG2a subtypes against tumor membrane proteins of B16 cells. In preventive vaccination, all treated groups showed delayed tumor growth. In addition, the group vaccinated to express only GM-CSF achieved 100% animal survival (P<0.005). Vaccination with GM-CSF+IL-12-producing B16 cells yielded lesser results (60% survival, P<0.005). Furthermore, all surviving animals remained disease-free after second tumor implantation 1 year later. The therapeutic vaccination strategies resulted in significantly delayed tumor growth, mainly using B16 cells producing GM-CSF+IL-12 cytokines, with 70% tumor growth inhibition (P<0.001)-although none of the animals reached overall survival. The results obtained suggest that the GM-CSF+IL-12 combination only increases the efficacy of therapeutic vaccines. No differences in classical regulatory T cells were found among the different groups.
Collapse
Affiliation(s)
- A Miguel
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Therapeutic cancer vaccines are an immunotherapy that targets tumor antigens to induce an active immune response. To date, Provenge® is the only therapeutic cancer vaccine approved by the United States Food and Drug Administration. Although therapeutic cancer vaccines have not been approved by the European Medicines Agency (EMA), they have been approved in several countries other than the United States (US) and the European Union (EU). Provenge® is the only approved cancer vaccine that showed significant primary endpoint efficacy in a phase III study at the time of approval. Retrospective analysis of 23 completed or terminated phase III studies showed that 74% (17/23) failed to demonstrate significant efficacy in the primary endpoint. The reasons for failure were surveyed in 13 of the 17 studies. Despite efforts to minimize tumor burden, including surgery and induction chemotherapy before therapeutic cancer vaccine therapy, 69% (9/13) of the phase III studies failed. These findings indicate that tumor burden may not be the only prognostic factor. Immunological response has often been used as a predictive factor, and a small number of sub-group analyses have succeeded in showing that immunological response is associated with the efficacy of therapeutic cancer vaccines. Being a prognostic factor, inclusion of immunological response in addition to tumor stage in the eligibility criteria or sub-group analysis may minimize study population heterogeneity, a key factor in the success of phase III studies.
Collapse
Affiliation(s)
- Chizuru Ogi
- Cooperative Major in Advanced Biomedical Sciences; Joint Graduate School of Tokyo Women's Medical University and Waseda University; Tokyo, Japan
| | | |
Collapse
|
20
|
Jones PC. Does a "thiol shield" protect tumors from natural IgM antibody, and, if so, how can it be suppressed? Med Hypotheses 2013; 80:425-30. [PMID: 23375413 DOI: 10.1016/j.mehy.2012.12.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 11/04/2012] [Accepted: 12/29/2012] [Indexed: 11/26/2022]
Abstract
Natural anti-tumor IgM antibodies are prevalent in the serum of cancer patients and normal subjects. Extensive research has been directed toward the ultimate goal of achieving a therapeutic effect from these antibodies either augmented by vaccination or by passive infusion. To date, the therapeutic effects have been limited. This thesis asserts that thiols within solid tumors reduce pentameric IgM to monomeric or other subunit form resulting in inactivation of its complement fixing and cross linking apoptosis inducing properties. A rationale for this normal physiological inactivation mechanism, possibly necessary for wound healing and pregnancy, is proposed along with therapeutic approaches, which would potentially suppress IgM inactivation.
Collapse
|
21
|
Comparative antitumor effect of preventive versus therapeutic vaccines employing B16 melanoma cells genetically modified to express GM-CSF and B7.2 in a murine model. Toxins (Basel) 2012. [PMID: 23202306 PMCID: PMC3509698 DOI: 10.3390/toxins4111058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cancer vaccines have always been a subject of gene therapy research. One of the most successful approaches has been working with genetically modified tumor cells. In this study, we describe our approach to achieving an immune response against a murine melanoma model, employing B16 tumor cells expressing GM-CSF and B7.2. Wild B16 cells were injected in C57BL6 mice to cause the tumor. Irradiated B16 cells transfected with GM-CSF, B7.2, or both, were processed as a preventive and therapeutic vaccination. Tumor volumes were measured and survival curves were obtained. Blood samples were taken from mice, and IgGs of each treatment group were also measured. The regulatory T cells (Treg) of selected groups were quantified using counts of images taken by confocal microscopy. Results: one hundred percent survival was achieved by preventive vaccination with the group of cells transfected with p2F_GM-CSF. Therapeutic vaccination achieved initial inhibition of tumor growth but did not secure overall survival of the animals. Classical Treg cells did not vary among the different groups in this therapeutic vaccination model.
Collapse
|
22
|
Carretero R, Wang E, Rodriguez AI, Reinboth J, Ascierto ML, Engle AM, Liu H, Camacho FM, Marincola FM, Garrido F, Cabrera T. Regression of melanoma metastases after immunotherapy is associated with activation of antigen presentation and interferon-mediated rejection genes. Int J Cancer 2012; 131:387-95. [PMID: 21964766 PMCID: PMC3504975 DOI: 10.1002/ijc.26471] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 09/12/2011] [Indexed: 01/11/2023]
Abstract
We present the results of a comparative gene expression analysis of 15 metastases (10 regressing and 5 progressing) obtained from 2 melanoma patients with mixed response following different forms of immunotherapy. Whole genome transcriptional analysis clearly indicate that regression of melanoma metastases is due to an acute immune rejection mediated by the upregulation of genes involved in antigen presentation and interferon mediated response (STAT-1/IRF-1) in all the regressing metastases from both patients. In contrast, progressing metastases showed low transcription levels of genes involved in these pathways. Histological analysis showed T cells and HLA-DR positive infiltrating cells in the regressing but not in the progressing metastases. Quantitative expression analysis of HLA-A,B and C genes on microdisected tumoral regions indicate higher HLA expression in regressing than in progressing metastases. The molecular signature obtained in melanoma rejection appeared to be similar to that observed in other forms of immune-mediated tissue-specific rejection such as allograft, pathogen clearance, graft versus host or autoimmune disease, supporting the immunological constant of rejection. We favor the idea that the major factor determining the success or failure of immunotherapy is the nature of HLA Class I alterations in tumor cells and not the type of immunotherapy used. If the molecular alteration is reversible by the immunotherapy, the HLA expression will be upregulated and the lesion will be recognized and rejected. In contrast, if the defect is structural the MHC Class I expression will remain unchanged and the lesion will progress.
Collapse
Affiliation(s)
- Rafael Carretero
- Departamento de Bioquímica, Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Departamento de Análisis Clínicos e Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | - Ana I. Rodriguez
- Departamento de Análisis Clínicos e Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| | - Jennifer Reinboth
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
- Department of Biochemistry, Biocenter, University of Wuerzburg, Wuerzburg, Germany
- Genelux Corporation, San Diego Science Center, San Diego, CA
| | - Maria L. Ascierto
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | - Alyson M. Engle
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | - Hui Liu
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | | | - Francesco M. Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | - Federico Garrido
- Departamento de Bioquímica, Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Departamento de Análisis Clínicos e Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| | - Teresa Cabrera
- Departamento de Bioquímica, Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Departamento de Análisis Clínicos e Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| |
Collapse
|
23
|
Madorsky-Rowdo FP, Lacreu ML, Mordoh J. Melanoma vaccines and modulation of the immune system in the clinical setting: building from new realities. Front Immunol 2012; 3:103. [PMID: 22566975 PMCID: PMC3343264 DOI: 10.3389/fimmu.2012.00103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 04/16/2012] [Indexed: 01/09/2023] Open
Abstract
To endow the immune system with the capacity to fight cancer has always attracted attention, although the clinical results obtained have been until recently disappointing. Cutaneous melanoma is a highly immunogenic tumor; therefore most of the attempts to produce cancer vaccines have been addressed to this disease. New advances in the comprehension of the mechanisms of antigen presentation by dendritic cells, in the immune responses triggered by adjuvants, as well as the understanding of the role of immunosuppressor molecules such as cytotoxic T-lymphocyte antigen-4 (CTLA-4), which led to the recent approval of the anti-CTLA-4 monoclonal antibody ipilimumab, have opened new hopes about the installment of immunotherapy as a new modality to treat cancer.
Collapse
|
24
|
Piérard GE. Cell proliferation in cutaneous malignant melanoma: relationship with neoplastic progression. ISRN DERMATOLOGY 2012; 2012:828146. [PMID: 22363864 PMCID: PMC3265211 DOI: 10.5402/2012/828146] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 11/30/2011] [Indexed: 12/12/2022]
Abstract
The establishment of the diagnosis of cutaneous malignant melanoma (CMM) always calls for histopathological confirmation. Further to the recognition of the CMM aspects, immunohistochemistry is helpful, in particular, in determining the size of the replicative compartment and the activity in each of the cell cycle phases (G(1), S, G(2), M). The involvement of cancer stem cells and transient amplifier cells in CMM genesis is beyond doubt. The proliferation activity is indicative of the neoplastic progression and is often related to the clinical growth rate of the neoplasm. It allows to distinguish high-risk CMM commonly showing a high growth rate, from those CMMs of lower malignancy associated with a more limited growth rate. The recruitment and progression of CMM cells in the cell cycle of proliferation depend on mitogen-activated protein kinase (MAPK) pathway and result from a loss of control normally involving a series of key regulatory cyclins. In addition, the apoptotic pathways potentially counteracting any excess in proliferative activity are out of the dependency of specific regulatory molecular mechanisms. Key molecular components involved in the deregulation of the growth fraction, the cell cycle phases of proliferation, and apoptosis are presently described in CMM.
Collapse
Affiliation(s)
- G. E. Piérard
- Department of Dermatopathology, University Hospital of Liège, 4000 Liège, Belgium
| |
Collapse
|
25
|
Abstract
BACKGROUND Immunotherapy for cutaneous malignancy involves manipulating the immune system to treat and prevent skin cancer. Although initial efforts were fraught with low success rates and technical challenges, more-recent endeavors have yielded response rates approaching 50% for treating metastatic melanoma. Many of these advances are a result of increasing knowledge of the immune system's intricacies and continued progress in laboratory techniques. OBJECTIVE To review our current understanding of the skin immune system and discuss how these factors contribute to the host response to malignancy and to report the current state of immunotherapeutic techniques. MATERIALS AND METHODS An extensive PubMed literature search was conducted in topics involving immunotherapy with specific relevance to cutaneous malignancy using the MeSH terms "immunotherapy" and "skin cancer." RESULTS Despite initially poor patient responses to these treatment modalities, recent gains in scientific knowledge and clinical intervention protocols have brought immunotherapy to the forefront of prospective skin cancer therapeutics, particularly for advanced melanoma. CONCLUSIONS Current treatment options for advanced cutaneous malignancies such as melanoma are low in efficacy. Immunotherapies have the potential to provide novel approaches to address this, particularly when used in combination. The authors have indicated no significant interest with commercial supporters.
Collapse
Affiliation(s)
- Sherrif F Ibrahim
- Division of Dermatologic Surgery, Department of Dermatology, University of Rochester Medical Center, Rochester, New York 14623, USA.
| | | | | |
Collapse
|
26
|
di Pietro A, Tosti G, Ferrucci PF, Testori A. The immunological era in melanoma treatment: new challenges for heat shock protein-based vaccine in the advanced disease. Expert Opin Biol Ther 2011; 11:1395-407. [PMID: 21801084 DOI: 10.1517/14712598.2011.605353] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Tumor-derived heat shock protein (HSP)-peptide complexes (HSPPCs) induced immunity against malignancies in preclinical trials, working across tumor types and bypassing the need to identify single immunogenic peptides. These results paved the way for the use of human gp96 obtained from autologous tumor samples as an anti-cancer vaccine. AREAS COVERED Autologous tumor-derived HSP gp96 peptide complex (HSPPC-96) vaccine is emerging as a tumor- and patient-specific cancer vaccine, with confirmed activity in several malignancies. It has been tested in Phase III clinical trials in advanced melanoma and kidney cancer with evidence for efficacy in patients with earlier stage disease. HSPPC-96-based vaccine demonstrated an excellent safety profile, thus emerging as a novel therapeutic approach with a suggestive role in cancer therapy. This review summarizes work on the use of HSPPC-96 as an autologous anti-tumor vaccine in advanced melanoma. Data were retrieved by PubMed and Medline research and using the authors' personal experience. EXPERT OPINION Further investigations are needed to understand the biological basis of immune functions in order to improve the clinical outcome of HSP-based cancer therapy. In the near future, the combination of HSP-based vaccines with other biological compounds might represent a successful strategy in the therapy of advanced melanoma.
Collapse
Affiliation(s)
- Alessandra di Pietro
- IEO, European Institute of Oncology, Melanoma Division, Via G. Ripamonti 435, 20141 Milan, Italy
| | | | | | | |
Collapse
|
27
|
Wieërs G, Demotte N, Godelaine D, van der Bruggen P. Immune suppression in tumors as a surmountable obstacle to clinical efficacy of cancer vaccines. Cancers (Basel) 2011; 3:2904-54. [PMID: 24212939 PMCID: PMC3759179 DOI: 10.3390/cancers3032904] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 07/01/2011] [Accepted: 07/07/2011] [Indexed: 02/07/2023] Open
Abstract
Human tumors are usually not spontaneously eliminated by the immune system and therapeutic vaccination of cancer patients with defined antigens is followed by tumor regressions only in a small minority of the patients. The poor vaccination effectiveness could be explained by an immunosuppressive tumor microenvironment. Because T cells that infiltrate tumor metastases have an impaired ability to lyse target cells or to secrete cytokine, many researchers are trying to decipher the underlying immunosuppressive mechanisms. We will review these here, in particular those considered as potential therapeutic targets. A special attention will be given to galectins, a family of carbohydrate binding proteins. These lectins have often been implicated in inflammation and cancer and may be useful targets for the development of new anti-cancer therapies.
Collapse
Affiliation(s)
- Grégoire Wieërs
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Nathalie Demotte
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Danièle Godelaine
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| | - Pierre van der Bruggen
- Ludwig Institute for Cancer Research and Université catholique de Louvain, de Duve Institute, 74 av. Hippocrate, P.O. Box B1-7403, B-1200 Brussels, Belgium; E-Mails: (G.W.); (N.D.); (D.G.)
| |
Collapse
|
28
|
Abstract
Immune-mediated cancer regression requires tumor infiltration by antigen-specific effector T cells, but lymphocytes are commonly sparse in melanoma metastases. Activated T cells express CXCR3, whose cognate chemokines are CXCL9/MIG, CXCL10/IP-10, and CXCL11/I-TAC. Little is known about expression of these chemokines in lymph node (LN) metastases of melanoma. We evaluated whether metastatic melanoma induces these CXCR3-cognate chemokines in human LN-derived tissues. In addition, as these chemokines can be induced by interferon (IFN), we evaluated whether type I or II IFNs (IFN-α or IFN-γ, respectively) can modulate chemokine expression in an in vitro model of the human tumor microenvironment. Production of CXCL9-11 by melanoma-infiltrated nodes (MIN) was no different than tumor-free nodes; both produced less chemokine than activated LN (sentinel immunized nodes, SIN). These data suggest that melanoma infiltration into LN neither induces nor reduces CXCL9-11. Stimulation with IFN-α or IFN-γ increased production of CXCL10-11 from MIN, but not tumor-free node or SIN. IFN-γ also increased production of CXCL9 in MIN. In IFN-treated SIN, CD14+ cells were the primary source of CXCL9-11, whereas melanoma cells were the source of chemokine in MIN. Melanoma cells in MIN express IFN receptors. Consistent with these observations, multiple human melanoma lines expressed IFN receptors and produced CXCL9-11 in response to IFN treatment. Thus, melanoma infiltration of LN is insufficient to induce the production of CXCL9-11, but melanoma may be a significant source of IFN-induced chemokines. Collectively, these data suggest that IFN-α or IFN-γ may act in the tumor microenvironment to increase the chemotactic gradient for CXCR3+ T cells.
Collapse
|
29
|
Kim SK, Wu X, Ragupathi G, Gathuru J, Koide F, Cheung NK, Panageas K, Livingston PO. Impact of minimal tumor burden on antibody response to vaccination. Cancer Immunol Immunother 2011; 60:621-7. [PMID: 21267719 PMCID: PMC3734789 DOI: 10.1007/s00262-011-0975-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 01/05/2011] [Indexed: 10/18/2022]
Abstract
Four randomized phase III trials conducted recently in melanoma patients in the adjuvant setting have been based in part on the correlation between antibody responses in immunized patients and improved survival. Each of these randomized trials demonstrated no clinical benefit, although again there was a significant correlation between antibody response after vaccination and disease free and overall survival. To better understand this paradox, we established a surgical adjuvant model targeting GD2 ganglioside on EL4 lymphoma cells injected into the foot pad followed by amputation at variable intervals. Our findings are (1) comparable strong therapeutic benefit resulted from treatment of mice after amputation with a GD2-KLH conjugate vaccine or with anti-GD2 monoclonal antibody 3F8. (2) The strongest correlation was between antibody induction in response to vaccination and prolonged survival. (3) Antibody titers in response to vaccination in tumor challenged mice as compared to unchallenged mice were far lower despite the absence of detectable recurrences at the time. (4) The half life of administered 3F8 monoclonal antibody (but not control antibody) in challenged mice administered was significantly shorter than the half life of 3F8 antibody in unchallenged controls. The correlation between vaccine-induced antibody titers and prolonged survival may reflect, at least in part, increased tumor burden in antibody-negative mice. Absorption of vaccine-induced antibodies by increased, although not detected tumor burden may also explain the correlation between vaccine-induced antibody titers and survival in the adjuvant clinical trials described above.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/biosynthesis
- Antibody-Dependent Cell Cytotoxicity
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Disease-Free Survival
- Enzyme-Linked Immunosorbent Assay
- Gangliosides/immunology
- Hemocyanins/immunology
- Immunoglobulin G/immunology
- Immunoglobulin G/therapeutic use
- Lymphoma/immunology
- Lymphoma/pathology
- Lymphoma/therapy
- Mice
- Mice, Inbred C57BL
- Tumor Burden
- Vaccination
- Vaccines, Conjugate/administration & dosage
- Vaccines, Conjugate/immunology
- Vaccines, Conjugate/therapeutic use
Collapse
Affiliation(s)
| | - Xiaohong Wu
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Govind Ragupathi
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | | | | - Nai-Kong Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Katherine Panageas
- Department of Biostatistics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Philip O. Livingston
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
30
|
Abstract
Malignant melanoma (MM) micrometastases are basically seen in three locations inside the peritumoral dermis. They are localized (i) inside the interstitial sector of the dermal stroma; (ii) abutted to the external surface of the microvasculature; and (iii) more rarely present inside vascular channels. Single-cell and paucicellular micrometastases may be disclosed using immunohistochemistry even in the absence of larger microsatellites, which represent micronodular nests of metastatic cells. The presence of microsatellites is frequently tied to markers of MM aggressiveness including thickness and the Ki-67 index. Micrometastases may be present in the same conditions, but even as early as thin MM showing a small growth fraction. Microsatellites as well as micrometastases appear to predict locoregional extension and decreased relapse-free interval, but not distant metastasis and overall survival. These considerations have implications for patient care since patients with microsatellites and micrometastases are now included in the clinical stage III category of the disease. Their implication as a prognostic factor is not fully dependent on or linked to other markers of MM aggressiveness.
Collapse
Affiliation(s)
- Pascale Quatresooz
- Department of Dermatopathology, University Hospital of Liège, Liège, Belgium
| | | |
Collapse
|
31
|
Un K, Kawakami S, Suzuki R, Maruyama K, Yamashita F, Hashida M. Suppression of Melanoma Growth and Metastasis by DNA Vaccination Using an Ultrasound-Responsive and Mannose-Modified Gene Carrier. Mol Pharm 2011; 8:543-54. [DOI: 10.1021/mp100369n] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Keita Un
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
- The Japan Society for the Promotion of Science (JSPS), Chiyoda-ku, Tokyo 102-8471, Japan
| | - Shigeru Kawakami
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Ryo Suzuki
- Department of Biopharmaceutics, School of Pharmaceutical Sciences, Teikyo University, 1091-1 Suwarashi, Midori-ku, Sagamihara, Kanagawa 252-5195, Japan
| | - Kazuo Maruyama
- Department of Biopharmaceutics, School of Pharmaceutical Sciences, Teikyo University, 1091-1 Suwarashi, Midori-ku, Sagamihara, Kanagawa 252-5195, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mitsuru Hashida
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Yoshida-ushinomiya-cho, Sakyo-ku, Kyoto 606-8302, Japan
| |
Collapse
|
32
|
Jorritsma A, Schumacher TNM, Haanen JBAG. Immunotherapeutic strategies: the melanoma example. Immunotherapy 2011; 1:679-90. [PMID: 20635992 DOI: 10.2217/imt.09.20] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
T-cell-based immunotherapy can be induced by nonspecific activation, by antigen-specific immunization, or by adoptive immunotherapy. In this review, progress in these areas is discussed as based on data from clinical trials for the treatment of metastatic melanoma. Nonspecific immunotherapy has been shown to result in low, but in some cases significant, levels of objective tumor responses, and is often associated with autoimmune reactions. Antigen-specific targeting of tumors via vaccination has only resulted in low to very low levels of objective responses, and these strategies seem to have most value when the T-cell repertoire is not affected by tolerance. Finally, adoptive immunotherapy can be applied by in vitro expansion of autologous lymphocytes that have escaped tolerance or by genetic transfer of allogeneic T-cell receptors (TCRs). Autologous adoptive T-cell transfer has resulted in a very high frequency of clinical responses when combined with chemotherapy and IL-2 administration in single-center studies. Although TCR gene transfer has, until now, only resulted in a low frequency of clinical responses, it does have a broader application potential, and optimization of this strategy is likely to improve its efficacy.
Collapse
Affiliation(s)
- Annelies Jorritsma
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
33
|
Abstract
Personalized vaccine, recognized after the failure of allogenic melanoma whole cell and lysate vaccine phase III trials, involves culturing cells from a patient's own tumor within a short duration and with less passages but with optimized expression of tumor-associated antigens (TAAs). Its feasibility is established by comparing pure cell lines generated from fresh and cryopreserved tissues (n=164) of patients with lymph node (LN) and distant metastases. Stable cell lines (from 67% of specimens) are subcultured after cryopreserving them. Pure cell lines established after eliminating fibroblasts (from 96% of the cell lines) include those from LN (69%), soft tissues including cutaneous (60%), liver (64%), lung (75%), bone (80%), brain (75%), and other sites (73%). Within 3.5 months, stable cell lines (> or =50 million cells) are established from initiating the cell culture. For LN metastases, the duration differs significantly (P2<0.05) between fresh (1.4-3.4 months) and cryopreserved (2.4-4.7 months) tissues. The expression of TAAs varies as follows: Tyrosinase (81%) >Melan-A (80%) >HMB45/gp-100 (75%) >Mel-5/TRP-1 (65%) >MAGE-1 (47%) > S-100 (28%). The number of TAAs per cell line differs between early (<7) and late (>7) passages. Among late passage cell lines, lesser percentage of cell lines express three to six antigens pointing out that early passage (<7) cell lines may be needed for antigen-targeted immunotherapy. This study provides a protocol for establishing cell lines within 2-5 months for personalized vaccine therapy for nodal and organ metastatic melanoma patients.
Collapse
|
34
|
Alexandrescu DT, Ichim TE, Riordan NH, Marincola FM, Di Nardo A, Kabigting FD, Dasanu CA. Immunotherapy for melanoma: current status and perspectives. J Immunother 2010; 33:570-90. [PMID: 20551839 PMCID: PMC3517185 DOI: 10.1097/cji.0b013e3181e032e8] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Immunotherapy is an important modality in the therapy of patients with malignant melanoma. As our knowledge about this disease continues to expand, so does the immunotherapeutic armamentarium. Nevertheless, successful preclinical models do not always translate into clinically meaningful results. The authors give a comprehensive analysis of most recent advances in the immune anti-melanoma therapy, including interleukins, interferons, other cytokines, adoptive immunotherapy, biochemotherapy, as well as the use of different vaccines. We also present the fundamental concepts behind various immune enhancement strategies, passive immunotherapy, as well as the use of immune adjuvants. This review brings into discussion the results of newer and older clinical trials, as well as potential limitations and drawbacks seen with the utilization of various immune therapies in malignant melanoma. Development of novel therapeutic approaches, along with optimization of existing therapies, continues to hold a great promise in the field of melanoma therapy research. Use of anti-CTLA4 and anti-PD1 antibodies, realization of the importance of co-stimulatory signals, which translated into the use of agonist CD40 monoclonal antibodies, as well as activation of innate immunity through enhanced expression of co-stimulatory molecules on the surface of dendritic cells by TLR agonists are only a few items on the list of recent advances in the treatment of melanoma. The need to engineer better immune interactions and to boost positive feedback loops appear crucial for the future of melanoma therapy, which ultimately resides in our understanding of the complexity of immune responses in this disease.
Collapse
Affiliation(s)
- Doru T Alexandrescu
- Division of Dermatology, University of California at San Diego, San Diego, CA, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Chhabra A. Mitochondria-centric activation induced cell death of cytolytic T lymphocytes and its implications for cancer immunotherapy. Vaccine 2010; 28:4566-72. [PMID: 20451638 PMCID: PMC3870157 DOI: 10.1016/j.vaccine.2010.04.074] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 04/15/2010] [Accepted: 04/23/2010] [Indexed: 12/23/2022]
Abstract
Premature death of the adoptively transferred cytolytic T lymphocytes (CTL) by means of activation induced cell death (AICD) represents one of the major constraints in devising an effective anti-cancer immune intervention strategy. Understanding the mechanism of AICD is, therefore, critical for developing methods to interfere with this death process. Although the existing paradigm on AICD centers around the initiation of the cascade of events originating from the engagement of death receptors leading to the activation of effector caspases and eventually resulting in cell death, recent findings have questioned the universal role of caspases as the cell death executioners. We here review our current understanding of the contribution of caspase-dependent and caspase-independent death executioners in AICD of T cells. We will also discuss the involvement of mitochondria-centric death pathway in AICD of human tumor associated antigen-specific primary CTL and its implications in cancer immunotherapy.
Collapse
Affiliation(s)
- Arvind Chhabra
- Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA.
| |
Collapse
|
36
|
Durrant LG, Pudney V, Spendlove I, Metheringham RL. Vaccines as early therapeutic interventions for cancer therapy: neutralising the immunosuppressive tumour environment and increasing T cell avidity may lead to improved responses. Expert Opin Biol Ther 2010; 10:735-48. [DOI: 10.1517/14712591003769790] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
37
|
Perez CA, Fu A, Onishko H, Hallahan DE, Geng L. Radiation induces an antitumour immune response to mouse melanoma. Int J Radiat Biol 2010; 85:1126-36. [DOI: 10.3109/09553000903242099] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
38
|
Ordaz ML, Larmonier N, Lybarger L. DC-expressed MHC class I single-chain trimer-based vaccines prime cytotoxic T lymphocytes against exogenous but not endogenous antigens. Cell Immunol 2010; 262:141-9. [PMID: 20199770 DOI: 10.1016/j.cellimm.2010.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 02/05/2010] [Indexed: 11/30/2022]
Abstract
The poor immunogenicity of many tumors can be partly explained by the inefficiency of the MHC class I peptide presentation pathway. MHC-I-based single-chain trimers (SCT) represent a new class of molecules with the potential to overcome this limitation. We here evaluated the ability of SCT presenting a melanoma antigen peptide (TRP-2) to prime cytotoxic T lymphocyte (CTL) responses in mice when given as DNA vaccines via Gene Gun or when expressed by dendritic cells. The SCT was unable to induce detectable priming or significant anti-tumor activity of CTL using either vaccination strategy, whereas control SCT (with an exogenous peptide) primed strong responses. This study thus provides the first data related to the use of SCT in combination with DC and their application toward self antigens and suggest this potent technology, alone, is insufficient to overcome self tolerance.
Collapse
Affiliation(s)
- Maria L Ordaz
- Department of Immunobiology, The University of Arizona, Tucson, AZ 85724, United States
| | | | | |
Collapse
|
39
|
Tosti G, di Pietro A, Ferrucci PF, Testori A. HSPPC-96 vaccine in metastatic melanoma patients: from the state of the art to a possible future. Expert Rev Vaccines 2010; 8:1513-26. [PMID: 19863242 DOI: 10.1586/erv.09.108] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Heat-shock proteins are highly conserved, stress-induced proteins with chaperone function for trafficking and delivering peptides within the different compartments of the cell. Tumor-derived heat-shock protein-peptide complexes (HSPPCs) can be used for vaccination against malignancies. In particular, the HSPPC-96-based vaccine vitespen (formerly Oncophage) is the first autologous cancer vaccine made from individual patients' tumors that has shown encouraging results in clinical trials. In Phase I and II clinical trials, this vaccine has shown activity on different malignancies, such as gastric cancer, colorectal cancer, pancreatic cancer, non-Hodgkin's lymphoma and chronic myelogenous leukemia. In Phase III clinical trials in melanoma and kidney cancer, it demonstrated an excellent safety profile with almost no toxicity. Heat-shock protein-based vaccines can be considered as a novel therapeutic approach with a promising role in cancer management.
Collapse
Affiliation(s)
- Giulio Tosti
- Division of Melanoma and Muscle-Cutaneous Sarcomas, IEO, European Institute of Oncology, Milan, Italy.
| | | | | | | |
Collapse
|
40
|
Abstract
Pediatric melanoma is rare but increasing in incidence. Because early diagnosis and treatment improves prognosis, clinicians need to include it as a possible diagnosis when evaluating a pigmented lesion in a pediatric patient. Some risk factors for melanoma include xeroderma pigmentosum, giant congenital melanocytic nevi, dysplastic nevus syndrome, atypical nevi, many acquired melanocytic nevi, family history of melanoma, and immunosuppression. Definitive treatment is with surgical excision. Adjuvant therapies such as chemotherapy, immunotherapy, and radiation therapy can be used in advanced cases.
Collapse
Affiliation(s)
- Melinda Jen
- Department of Dermatology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | | | | |
Collapse
|
41
|
|
42
|
Abstract
Metastatic dormancy of melanoma has not received sufficient attention, most likely because once detectable, metastasis is almost invariably fatal and, understandably, the focus has been on finding ways to prolong life of patients with overt recurrences. Nevertheless, analysis of the published clinical and experimental data on melanoma indicates that some aspect of melanoma biology imitate traits recently associated with dormancy in other solid cancers. Among them the ability of some melanomas to disseminate early during primary tumor progression and once disseminated, to remain undetected (dormant) for years. Comparison of cutaneous and uveal melanoma indicates that, in spite of being of the same origin, they differ profoundly in their clinical progression. Importantly for this discussion, between 40 and 50% of uveal melanoma remain undetected for longer than a decade, while less than 5% of cutaneous melanoma show this behavior. Both types of melanoma have activating oncogene mutations that provide autonomous pro-proliferative signals, yet the consensus is that those are not sufficient for tumor progression. If that is the case, it is possible to envision that signals from outside the tumor cell, (microenvironment) shape the fate of an individual disseminated cell, regardless of an oncogene mutation, to progress or to pause in a state of dormancy. To stimulate further debate and inquiry we describe here a few examples of potential signals that might modify the fate of disseminated cell and provide brief description of the current knowledge on dormancy in other cancers. Our hope is to convince the reader that disseminated melanoma cells do enter periods of prolonged dormancy and that finding ways to induce it, or to prolong it, might mean an extension of symptoms-free life for melanoma patients. Ultimately, understanding the biology of dormancy and the mechanisms of dormant cell survival, might allow for their specific targeting and elimination.
Collapse
Affiliation(s)
- Liliana Ossowski
- Division of Hematology and Oncology, Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA.
| | | |
Collapse
|
43
|
Gridelli C, Rossi A, Maione P, Ferrara ML, Castaldo V, Sacco PC. Vaccines for the treatment of non-small cell lung cancer: a renewed anticancer strategy. Oncologist 2009; 14:909-20. [PMID: 19726457 DOI: 10.1634/theoncologist.2009-0017] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Carcinoma of the lung is the leading cause of cancer death worldwide, with non-small cell lung cancer (NSCLC) constituting about 85% of all new diagnoses. Standard approaches for each NSCLC stage have reached a plateau in effectiveness. A variety of novel approaches are now being investigated to improve the outcome of this disease. Despite decades of research, no specific active cancer vaccine has, to date, been approved for NSCLC therapy; nevertheless, vaccine therapy has recently re-emerged as a potential therapeutic approach. In particular, several new paradigms have stemmed from recent clinical findings both in the use of combination therapy approaches with more sophisticated specific vaccines and in clinical trial design and endpoint analyses. Several vaccine therapies have been investigated in NSCLC, including in the early and advanced disease stages. The best results appear to be in the adjuvant settings and in locally advanced NSCLC. In fact, in these two settings, phase III randomized trials are ongoing evaluating the melanoma-associated antigen A3 vaccine and the liposomal BLP25 vaccine. This paper reviews the main clinical trials involving several different cancer vaccines employed in the treatment of early and advanced stage NSCLC, focusing on those in advanced stages of development.
Collapse
Affiliation(s)
- Cesare Gridelli
- Division of Medical Oncology, S.G. Moscati Hospital, Avellino, Italy.
| | | | | | | | | | | |
Collapse
|
44
|
Sharma A, Czerniecki BJ. Developing dendritic cell-based therapies to condition immune responses to novel oncogenic proteins and stem cells. Expert Rev Clin Pharmacol 2009; 2:517-26. [PMID: 22112225 DOI: 10.1586/ecp.09.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cancer vaccines have been disappointing when utilized as stand-alone therapy, especially in late disease settings. However, recent clinical studies in prostate cancer have suggested that dendritic cellular (DC) vaccines may impact patient survival, reviving the notion that cancer vaccines can impact established cancer. In this review we will highlight the advances that have been made in the development of DC-based therapies activated by Toll-like receptor agonists with the capacity to condition toward strong Th1 cellular responses, through the production of cytokines and chemokines, and a capacity to induce apoptosis of tumor cells. Used in early cancer settings, these DCs induce clinically effective immune responses, thus shifting the emphasis toward using these cells earlier in the disease process. We will also discuss targeting novel molecules and cancer stem cells that can eliminate cells with high metastatic potential, moving DC-based therapies into mainstream cancer therapy.
Collapse
Affiliation(s)
- Anupama Sharma
- Research and Department of Surgery, Rena Rowan Breast Center, Abramson Cancer Center, PENN Medicine, University of Pennsylvania, PA, USA.
| | | |
Collapse
|
45
|
Mansfield AS, Markovic SN. Novel therapeutics for the treatment of metastatic melanoma. Future Oncol 2009; 5:543-57. [PMID: 19450181 DOI: 10.2217/fon.09.15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Metastatic malignant melanoma is an incurable disease with a median survival of 8.5 months and a probability of surviving 5 years after the diagnosis of less than 5%. To date, no systemic therapy has meaningfully changed these survival end points. Currently, in the USA the FDA has approved three agents for the treatment of metastatic melanoma: hydroxyurea, dacarbazine and interleukin-2. None of these have demonstrated a meaningfully prolonged survival of patients with metastatic melanoma. Therefore, a number of innovative therapeutic strategies have been pursued to improve outcomes, including immune therapy, tyrosine kinase inhibitors and angiogenesis inhibitors. Herein, we review some of the recent advances in novel therapeutic developments for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Aaron S Mansfield
- Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | |
Collapse
|
46
|
Immunotherapy for malignant melanoma--tracing Ariadne's thread through the labyrinth. Eur J Cancer 2009; 45:2266-73. [PMID: 19497734 DOI: 10.1016/j.ejca.2009.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Revised: 04/28/2009] [Accepted: 05/01/2009] [Indexed: 12/18/2022]
Abstract
A working group (FEBIM) within the European Organisation for Research and Treatment of Cancer undertook extensive studies on the possible association of infectious diseases and the risk of malignant melanoma. These studies provided evidence that several infectious diseases and also some vaccines including the anti-tuberculosis vaccine, BCG, derived from Mycobacterium bovis, confer a significant level of protection against this form of cancer. In recent years, the importance of immunoregulatory networks in the establishment of tolerance to tumour antigens and the key role of the innate immune system in the development of such networks have been recognised. The molecular patterns of micro-organisms activate pattern recognition receptors on antigen presenting cells and determine the qualitative nature of the ensuing immune response. Bacteria in the actinomycetales family, notably members of the genus Mycobacterium, exhibit particularly powerful adjuvant activity and profoundly affect underlying patterns of immune reactivity. In particular, there is growing evidence that a heat-killed preparation of a strain of Mycobacterium vaccae is able to down-regulate patterns of immune reactivity that favour the tumour and to induce those that lead to anti-cancer immune responses. The results of preliminary clinical observations with melanoma patients, and published studies on other cancers, point to the need for more formal clinical trials.
Collapse
|
47
|
di Pietro A, Tosti G, Ferrucci PF, Testori A. Oncophage: step to the future for vaccine therapy in melanoma. Expert Opin Biol Ther 2009; 8:1973-84. [PMID: 18990084 DOI: 10.1517/14712590802517970] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Heat-shock proteins (HSPs) are a group of proteins whose expression is increased when the cells are exposed to elevated temperatures or other stressful conditions. This increase in expression is transcriptionally regulated. The function of HSPs is similar in virtually all living organisms, from bacteria to humans. Their expression also occur under non-stressful conditions, simply 'monitoring' the cell's proteins, i.e., they carry old proteins to the cell's 'recycling bin' and they help newly synthesized proteins fold properly. These activities are part of a cell's own repair system. HSPs are molecular chaperones for protein molecules. They are usually cytoplasmic proteins and they perform functions in various intracellular processes. Tumour-derived HSP-peptide complexes (HSPPCs) can be used for vaccination against malignancies. In particular, HSPPC-96 complex, called Vitespen (formerly Oncophage) is a HSPs-based vaccine made from individual patients' tumours with a promising role in cancer management. This vaccine has been extensively studied in Phase I and II clinical trials, showing activity on different malignancies, including gastric cancer, colorectal cancer, pancreatic cancer, non-Hodgkin's lymphoma and chronic myelogenous leukaemia. The vaccine has also been studied in Phase III clinical trials in melanoma and kidney cancer, showing an excellent safety profile with essentially no toxicity. Thus, HSP-based vaccines are a novel therapeutic approach with a promising role in cancer management.
Collapse
Affiliation(s)
- Alessandra di Pietro
- Division of Melanoma and Musculo-Cutaneous Sarcomas, IEO-European Institute of Oncology, 435, Via Giuseppe Ripamonti, 20141 Milan, Italy
| | | | | | | |
Collapse
|
48
|
Gaymalov ZZ, Yang Z, Pisarev VM, Alakhov VY, Kabanov AV. The effect of the nonionic block copolymer pluronic P85 on gene expression in mouse muscle and antigen-presenting cells. Biomaterials 2009; 30:1232-45. [PMID: 19064283 PMCID: PMC2667955 DOI: 10.1016/j.biomaterials.2008.10.064] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Accepted: 10/31/2008] [Indexed: 11/29/2022]
Abstract
DNA vaccines can be greatly improved by polymer agents that simultaneously increase transgene expression and activate immunity. We describe here Pluronic P85 (P85), a triblock copolymer of ethylene oxide (EO) and propylene oxide (PO) EO(26)-PO(40)-EO(26). Using a mouse model we demonstrate that co-administration of a bacterial plasmid DNA with P85 in a skeletal muscle greatly increases gene expression in the injection site and distant organs, especially the draining lymph nodes and spleen. The reporter expression colocalizes with the specific markers of myocytes and keratinocytes in the muscle, as well as dendritic cells (DCs) and macrophages in the muscle, lymph nodes and spleen. Furthermore, DNA/P85 and P85 alone increase the systemic expansion of CD11c+ (DC), and local expansion of CD11c+, CD14+ (macrophages) and CD49b+ (natural killer) cell populations. DNA/P85 (but not P85) also increases maturation of local DC (CD11c+ CD86+, CD11c+ CD80 +, and CD11c+ CD40+. We suggest that DNA/P85 promotes the activation and recruitment of the antigen-presenting cells, which further incorporate, express and carry the transgene to the immune system organs.
Collapse
Affiliation(s)
- Zagit Z. Gaymalov
- Center for Drug Delivery and Nanomedicine and Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, United States
| | - Zhihui Yang
- Center for Drug Delivery and Nanomedicine and Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, United States
| | - Vladimir M. Pisarev
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805
| | - Valery Yu. Alakhov
- Supratek Pharma Inc., 215 Bvd. Bouchard, Suite 1315, Dorval, Quebec H9S1A9, Canada
| | - Alexander V. Kabanov
- Center for Drug Delivery and Nanomedicine and Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, United States
- Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119899 Moscow, Russia
| |
Collapse
|
49
|
Abstract
Over the last century, vaccine studies have demonstrated that the human immune system, with appropriate help, can limit or prevent infection against otherwise lethal pathogens. Encouraged by these results, success in animal models and numerous well-documented reports of immune-mediated melanoma regression in humans, investigators developed melanoma vaccines. However, despite considerable laboratory evidence for vaccine-induced immune responses, clinical responses remain poor. Recent studies have elucidated several mechanisms that hinder or prevent the creation of successful vaccines and suggest novel approaches to overcome these barriers. Unraveling the mechanisms of autoimmunity, dendritic cell activation, regulatory T cells and Toll-like receptors will generate novel vaccines that, when used in conjunction with standard adjuvant therapies, may result in improved clinical outcomes. The objective of this review is to provide an overall summary of recent clinical trials with melanoma vaccines and highlight novel vaccine strategies to evaluate in the near future.
Collapse
Affiliation(s)
- Lee B Riley
- St Luke's Cancer Center, St Luke's Hospital and Health Network, 801 Ostrum Street, Bethlehem, PA 18015, USA.
| | | |
Collapse
|
50
|
Abstract
For many years, various cancer vaccines have been widely evaluated, however clinical responses remain rare. In this review, we attempt to address the question of which delivery strategies and platforms are feasible to produce clinical response and define the characteristics of the strategy that will induce long-lasting antitumor response. We limit our analysis and discussion to microparticles/nanoparticles, liposomes, heat-shock proteins, viral vectors and different types of adjuvants. This review aims to provide an overview of the specific characteristics, strengths and limitations of these delivery systems, focusing on their impacts on the development of melanoma vaccine. To date, only adoptive T-cell transfer has shown promising clinical outcomes compared to other treatments.
Collapse
Affiliation(s)
- Yin Hwa Lai
- Department of Pharmaceutical Sciences, Mercer University, College of Pharmacy and Health Sciences, Atlanta, GA 30341, USA.
| | | |
Collapse
|