1
|
Farasatkia A, Maeso L, Gharibi H, Dolatshahi-Pirouz A, Stojanovic GM, Edmundo Antezana P, Jeong JH, Federico Desimone M, Orive G, Kharaziha M. Design of nanosystems for melanoma treatment. Int J Pharm 2024; 665:124701. [PMID: 39278291 DOI: 10.1016/j.ijpharm.2024.124701] [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: 06/25/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Melanoma is a prevalent and concerning form of skin cancer affecting millions of individuals worldwide. Unfortunately, traditional treatments can be invasive and painful, prompting the need for alternative therapies with improved efficacy and patient outcomes. Nanosystems offer a promising solution to these obstacles through the rational design of nanoparticles (NPs) which are structured into nanocomposite forms, offering efficient approaches to cancer treatment procedures. A range of NPs consisting of polymeric, metallic and metal oxide, carbon-based, and virus-like NPs have been studied for their potential in treating skin cancer. This review summarizes the latest developments in functional nanosystems aimed at enhancing melanoma treatment. The fundamentals of these nanosystems, including NPs and the creation of various functional nanosystem types, facilitating melanoma treatment are introduced. Then, the advances in the applications of functional nanosystems for melanoma treatment are summarized, outlining both their benefits and the challenges encountered in implementing nanosystem therapies.
Collapse
Affiliation(s)
- Asal Farasatkia
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Lidia Maeso
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Hamidreza Gharibi
- Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | | | - Goran M Stojanovic
- Department of Electronics, Faculty of Technical Sciences, University of Novi Sad, 21000, Novi Sad, Serbia
| | - Pablo Edmundo Antezana
- Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA, CONICET), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Jee-Heon Jeong
- Laboratory of Drug Delivery and Cell Therapy (LDDCT). Department of Precision Medicine. School of Medicine, Sungkyunkwan University. South Korea
| | - Martin Federico Desimone
- Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA, CONICET), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina; Instituto de Ciências Biológicas (ICB), Universidade Federal do Rio Grande - FURG, Rio Grande, RS, Brazil
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria 01007, Spain.
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| |
Collapse
|
2
|
Chen F, Sheng J, Li X, Gao Z, Hu L, Chen M, Fei J, Song Z. Tumor-associated macrophages: orchestrators of cholangiocarcinoma progression. Front Immunol 2024; 15:1451474. [PMID: 39290697 PMCID: PMC11405194 DOI: 10.3389/fimmu.2024.1451474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a rare but highly invasive cancer, with its incidence rising in recent years. Currently, surgery remains the most definitive therapeutic option for CCA. However, similar to other malignancies, most CCA patients are not eligible for surgical intervention at the time of diagnosis. The chemotherapeutic regimen of gemcitabine combined with cisplatin is the standard treatment for advanced CCA, but its effectiveness is often hampered by therapeutic resistance. Recent research highlights the remarkable plasticity of tumor-associated macrophages (TAMs) within the tumor microenvironment (TME). TAMs play a crucial dual role in either promoting or suppressing tumor development, depending on the factors that polarize them toward pro-tumorigenic or anti-tumorigenic phenotypes, as well as their interactions with cancer cells and other stromal components. In this review, we critically examine recent studies on TAMs in CCA, detailing the expression patterns and prognostic significance of different TAM subtypes in CCA, the mechanisms by which TAMs influence CCA progression and immune evasion, and the potential for reprogramming TAMs to enhance anticancer therapies. This review aims to provide a framework for deeper future research.
Collapse
Affiliation(s)
- Fei Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jian Sheng
- Department of Research and Teaching, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaoping Li
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhaofeng Gao
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lingyu Hu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Minjie Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jianguo Fei
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhengwei Song
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
3
|
Yang C, Lei J, Kang X, Zhang P, Zheng S, Li Q, Zhang J. A Yeast Cell Wall Derived Hybrid Hydrogel with Photothermal and Immune Combined Modality Therapy for Enhanced Anti-Melanoma Efficacy. Int J Nanomedicine 2023; 18:5423-5440. [PMID: 37767196 PMCID: PMC10520258 DOI: 10.2147/ijn.s409674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Introduction The effect of traditional treatment for melanoma is quite limited, especially for its recurrence. As the major components of yeast cell wall, chitin and β-glucan exhibit good immune activation effect and are promising candidates for adjuvant. Therefore, melanoma cell membrane (CM) and indocyanine green (ICG) was loaded in a chitin and β-glucan hybrid hydrogel to achieve an enhanced anti-melanoma therapy. Methods The novel hybrid hydrogel was prepared, and its physicochemical properties were examined. Its effect towards melanoma prevention and treatment was evaluated via a melanoma-bearing mice model. Results The CM-ICG-hybrid hydrogel was successfully prepared with excellent injectability, self-healing, drug loading, rheological, in vitro and in vivo photothermal stability, and retention properties. It also exhibited good cellular and in vivo safety profiles. In the primary melanoma mice model, it quickly ablated the in-situ melanoma, effectively inhibited the tumor growth, increased the survival rate of melanoma-bearing mice, and increased the level of IFN-γ and TNF-α. In the distal secondary melanoma model, it efficiently prevented the reoccurrence of melanoma and activated the memory T cells. In both models, a synergistic effect of photothermal therapy and immune therapy was found. The hydrogel effectively recruited CD3+ CD4+ T cells and CD3+ CD8+ T cells, inhibited the proliferation of melanoma cells, and induced the apoptosis of melanoma cells. Conclusion The hybrid hydrogel was successfully prepared, and it showed excellent efficacy towards melanoma prevention and treatment due to its efficient tumor ablation and immune activation capability.
Collapse
Affiliation(s)
- Chen Yang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jiaxing Lei
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Ximeng Kang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Peipei Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Shaohua Zheng
- The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Qingqing Li
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| |
Collapse
|
4
|
Fobian SF, Cheng Z, ten Hagen TLM. Smart Lipid-Based Nanosystems for Therapeutic Immune Induction against Cancers: Perspectives and Outlooks. Pharmaceutics 2021; 14:26. [PMID: 35056922 PMCID: PMC8779430 DOI: 10.3390/pharmaceutics14010026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy, a promising and widely applied mode of oncotherapy, makes use of immune stimulants and modulators to overcome the immune dysregulation present in cancer, and leverage the host's immune capacity to eliminate tumors. Although some success has been seen in this field, toxicity and weak immune induction remain challenges. Liposomal nanosystems, previously used as targeting agents, are increasingly functioning as immunotherapeutic vehicles, with potential for delivery of contents, immune induction, and synergistic drug packaging. These systems are tailorable, multifunctional, and smart. Liposomes may deliver various immune reagents including cytokines, specific T-cell receptors, antibody fragments, and immune checkpoint inhibitors, and also present a promising platform upon which personalized medicine approaches can be built, especially with preclinical and clinical potentials of liposomes often being frustrated by inter- and intrapatient variation. In this review, we show the potential of liposomes in cancer immunotherapy, as well as the methods for synthesis and in vivo progression thereof. Both preclinical and clinical studies are included to comprehensively illuminate prospects and challenges for future research and application.
Collapse
Affiliation(s)
| | | | - Timo L. M. ten Hagen
- Laboratory Experimental Oncology (LEO), Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (S.-F.F.); (Z.C.)
| |
Collapse
|
5
|
Liu S, Zhang J, Fang S, Zhang Q, Zhu G, Tian Y, Zhao M, Liu F. Macrophage polarization contributes to the efficacy of an oncolytic HSV-1 targeting human uveal melanoma in a murine xenograft model. Exp Eye Res 2021; 202:108285. [PMID: 33039456 DOI: 10.1016/j.exer.2020.108285] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 09/04/2020] [Accepted: 09/29/2020] [Indexed: 12/28/2022]
Abstract
Uveal melanoma (UM), the most common primary malignant tumor of the eye in adults, is difficult-to-treat. UM has a relatively high mortality secondary to distant metastasis and poor overall survival with existing therapies. The oncolytic virus herpes simplex virus type-1 (HSV-1) has been approved for clinical use in melanoma. This double-stranded DNA virus was suspected to directly activate lysis specifically in neoplastic cells. We tested the antitumor efficacy of recombinant oncolytic HSV-1-EGFP (oHSV-EGFP) in UM and characterized the local and systemic antitumor innate immune response in a murine xenograft model. We first determined the efficacy of the oncolytic virus in 92.1, MUM2B and MP41 cell lines. In murine xenograft models, oHSV-EGFP reduced intraocular tumors as well as systemic subcutaneous tumors. A significant change in cytokines was observed in viral infected cells, especially a rise in IFNγ. In vivo analyses showed increased anti-tumorigenic M1 macrophages and decreased pro-tumorigenic M2 macrophages in peripheral blood, and intraocular and distant tumors after intravitreal viral treatment. Increased infiltration of natural killer cells and mature dendritic cells was also detected after viral treatment. In addition, no virus was detected in major organs after the treatment. Our data support that oHSV-EGFP is effective, neoplasm specific, immune active and safe, providing possible clinical translatable options to treat ocular and metastatic UM.
Collapse
Affiliation(s)
- Sisi Liu
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Guidong Zhu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Yifu Tian
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China.
| |
Collapse
|
6
|
Gargett T, Abbas MN, Rolan P, Price JD, Gosling KM, Ferrante A, Ruszkiewicz A, Atmosukarto IIC, Altin J, Parish CR, Brown MP. Phase I trial of Lipovaxin-MM, a novel dendritic cell-targeted liposomal vaccine for malignant melanoma. Cancer Immunol Immunother 2018; 67:1461-1472. [PMID: 30014244 PMCID: PMC11028356 DOI: 10.1007/s00262-018-2207-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 07/09/2018] [Indexed: 01/10/2023]
Abstract
INTRODUCTION In this phase I study using a 3 + 3 dose escalation design, the safety, dose-limiting toxicity (DLT), immunogenicity and efficacy of intravenous Lipovaxin-MM-a multi-component dendritic cell-targeted liposomal vaccine against metastatic melanoma-was investigated. METHODS Twelve subjects with metastatic cutaneous melanoma were recruited in three cohorts. Patients in Cohort A (n = 3) and Cohort B (n = 3) received three doses of 0.1 and 1 mL of Lipovaxin-MM, respectively, every 4 weeks. Patients in Cohort C (n = 6) received four doses of 3 mL vaccine weekly. Immunologic assessments of peripheral blood were made at regular intervals and included leukocyte subsets, cytokine levels, and Lipovaxin-MM-specific T-cell and antibody reactivities. Tumor responses were assessed by RECIST v1.0 at screening, then 8 weekly in Cohorts A and B and 6 weekly in Cohort C. RESULTS Of a total of 94 adverse events (AEs) reported in ten subjects, 43 AEs in six subjects were considered to be possibly or probably vaccine-related. Most (95%) vaccine-related AEs were grade 1 or 2, two (5%) grade 3 vaccine-related AEs of anemia and lethargy were recorded, and higher grade AEs and DLTs were not observed. No consistent evidence of vaccine-specific humoral or cellular immune responses was found in post-immunization blood samples. One patient had a partial response, two patients had stable disease, and the remaining patients had progressive disease. CONCLUSIONS Lipovaxin-MM was well tolerated and without clinically significant toxicity. Immunogenicity of Lipovaxin-MM was not detected. Partial response and stable disease were observed in one and two patients, respectively.
Collapse
Affiliation(s)
- Tessa Gargett
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - M Nazim Abbas
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Paul Rolan
- School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | | | | | - Antonio Ferrante
- School of Medicine, The University of Adelaide, Adelaide, SA, Australia
- Department of Immunopathology, SA Pathology, Women's and Children's Hospital, Adelaide, SA, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Andrew Ruszkiewicz
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
- Division of Anatomical Pathology, SA Pathology, Adelaide, SA, Australia
| | | | - Joseph Altin
- Division of Biomedical Science and Biochemistry, Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, the John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Michael P Brown
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, Australia.
- School of Medicine, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
7
|
Barik S, Banerjee S, Sarkar M, Bhuniya A, Roy S, Bose A, Baral R. Neem leaf glycoprotein optimizes effector and regulatory functions within tumor microenvironment to intervene therapeutically the growth of B16 melanoma in C57BL/6 mice. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.trivac.2013.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
8
|
Kawahara M, Takaku H. Intradermal immunization with combined baculovirus and tumor cell lysate induces effective antitumor immunity in mice. Int J Oncol 2013; 43:2023-30. [PMID: 24101126 DOI: 10.3892/ijo.2013.2125] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 09/13/2013] [Indexed: 11/06/2022] Open
Abstract
Although tumor lysate contains all the potential helper and killer epitopes capable of stimulating T cells, it is difficult to use as a cancer vaccine because it suppresses dendritic cell (DC) function. We report that wild-type baculovirus possesses an adjuvant effect to improve the immunogenicity of tumor lysate. When mice were administered CT26 tumor cell lysate combined with baculovirus intradermally, antitumor immunity was induced and rejection of CT26 tumor growth was observed in 40% of the immunized mice. In contrast, such antitumor immunity was not elicited in mice inoculated with tumor cell lysate or baculovirus alone. In tumor-bearing mice, which had previously received the combined baculovirus and tumor lysate vaccine, the established tumors were completely eradicated by administering a booster dose of the combined vaccine. This antitumor effect was attributed to tumor-specific T cell immunity mediated primarily by CD8⁺ T cells. Baculovirus also strongly activated DCs loaded with tumor lysate. Increased interleukin (IL)-6 and IL-12p70 production were also observed in DCs co-cultured with tumor cell lysate and baculovirus. Our study demonstrates that combined baculovirus and tumor lysate vaccine can effectively stimulate DCs to induce acquired antitumor immunity.
Collapse
Affiliation(s)
- Mamoru Kawahara
- Research and Development Department, Japan BCG Laboratory, Kiyose, Tokyo 204-0022, Japan
| | | |
Collapse
|
9
|
Hooper M, Wenkert D, Bitman B, Dias VC, Bartley Y. Malignancies in children and young adults on etanercept: summary of cases from clinical trials and post marketing reports. Pediatr Rheumatol Online J 2013; 11:35. [PMID: 24225257 PMCID: PMC3851136 DOI: 10.1186/1546-0096-11-35] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 09/19/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Malignancy risk may be increased in chronic inflammatory conditions that are mediated by tumor necrosis factor (TNF), such as juvenile idiopathic arthritis (JIA), but the role of TNF in human cancer biology is unclear. In response to a 2011 United States Food & Drug Administration requirement of TNF blocker manufacturers, we evaluated reporting rates of all malignancies in patients =30 years old who received the TNF blocker etanercept. METHODS All malignancies in etanercept-exposed patients aged =30 years from the Amgen clinical trial database (CTD) and postmarketing global safety database (PMD) were reviewed. PMD reporting rates were generated using exposure information based on commercial sources. Age-specific incidence rates of malignancy for the general US population were generated from the Surveillance Epidemiology and End Results (SEER) database v7.0.9. RESULTS There were 2 malignancies in the CTD: 1 each in etanercept and placebo/comparator arms (both in patients 18-30 years old). Postmarketing etanercept exposure was 231,404 patient-years (62,379 patient-years in patients 0-17 years; 168,485 patient-years in patients 18-30 years). Reporting rates of malignancy per 100,000 patient-years in the PMD and incidence rates in SEER were 32.0 and 15.9, respectively, for patients 0-17 years and 46.9 and 42.1 for patients 18-30 years old. Reporting rates were higher than SEER incidence rates for Hodgkin lymphoma in the 0-17 years age group. PMD reporting rates per 100,000 patient-years and SEER incidence rates per 100,000 person-years for Hodgkin lymphoma were 9.54 and 0.9, respectively, for patients 0-17 years and 1.8 and 4.2 for patients 18-30 years old. There were =5 cases of leukemia, lymphoma, melanoma, thyroid, and cervical cancers. Leukemia, non-Hodgkin lymphoma, melanoma, thyroid cancer, and cervical cancer rates were similar in the PMD and SEER. CONCLUSIONS Overall PMD malignancy reporting rates in etanercept-treated patients 0-17 years appeared higher than incidence rates in SEER, attributable to rates of Hodgkin lymphoma. Comparison to patients with similar burden of disease cannot be made; JIA, particularly very active disease, may be a risk factor for lymphoma. No increased malignancy reporting rate in the PMD relative to SEER was observed in the young-adult age group.
Collapse
Affiliation(s)
- Michele Hooper
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, USA
| | | | - Bojena Bitman
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, USA
| | - Virgil C Dias
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, USA
| | | |
Collapse
|
10
|
Bendle GM, Linnemann C, Bies L, Song JY, Schumacher TNM. Blockade of TGF-β Signaling Greatly Enhances the Efficacy of TCR Gene Therapy of Cancer. THE JOURNAL OF IMMUNOLOGY 2013; 191:3232-9. [DOI: 10.4049/jimmunol.1301270] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
11
|
Buhrman JD, Slansky JE. Improving T cell responses to modified peptides in tumor vaccines. Immunol Res 2013; 55:34-47. [PMID: 22936035 DOI: 10.1007/s12026-012-8348-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Immune recognition and elimination of cancerous cells is the primary goal of cancer immunotherapy. However, obstacles including immune tolerance and tumor-induced immunosuppression often limit beneficial immune responses. Vaccination is one proposed intervention that may help to overcome these issues and is an active area of study in cancer immunotherapy. Immunizing with tumor antigenic peptides is a promising, straight-forward vaccine strategy hypothesized to boost preexisting antitumor immunity. However, tumor antigens are often weak T cell agonists, attributable to several mechanisms, including immune self-tolerance and poor immunogenicity of self-derived tumor peptides. One strategy for overcoming these mechanisms is vaccination with mimotopes, or peptide mimics of tumor antigens, which alter the antigen presentation and/or T cell activation to increase the expansion of tumor-specific T cells. Evaluation of mimotope vaccine strategies has revealed that even subtle alterations in peptide sequence can dramatically alter antigen presentation and T cell receptor recognition. Most of this research has been performed using T cell clones, which may not be accurate representations of the naturally occurring antitumor response. The relationship between clones generated after mimotope vaccination and the polyclonal T cell repertoire is unclear. Our work with mimotopes in a mouse model of colon carcinoma has revealed important insights into these issues. We propose that the identification of mimotopes based on stimulation of the naturally responding T cell repertoire will dramatically improve the efficacy of mimotope vaccination.
Collapse
Affiliation(s)
- Jonathan D Buhrman
- Integrated Department of Immunology, University of Colorado School of Medicine, National Jewish Health, Denver, CO 80206, USA
| | | |
Collapse
|
12
|
Characterization of blood monocyte phenotype in patients with endometrial cancer. Int J Gynecol Cancer 2013; 22:1500-8. [PMID: 23051953 DOI: 10.1097/igc.0b013e3182249273] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Endometrial tumors induce various tumor escape mechanisms that result in immunosuppression in patients and, ultimately, tumor progression. Blood monocytes are able to exhibit potent cytotoxic action against tumor cells where novel immunotherapeutics targeting antigen-presenting cells including dendritic cells, and blood monocytes are being used as a means of delivering immunogens to stimulate an antitumor and, ultimately, therapeutic response. This study shows that peripheral blood monocytes from patients with endometrial cancer show functional deficiencies, and these deficiencies can be characterized by phenotypic changes as well as altered cytokine secretion. METHODS This study assessed the phenotypic changes of peripheral blood monocytes by flow cytometry as well as the functional status via cytokine production measured by enzyme-linked immunosorbent assay in patients with endometrial cancer versus controls. RESULTS Altered blood monocyte phenotype incorporating a decrease in costimulatory and adhesion factor expression and increased expression of vascular endothelial growth factor receptor 1 in patients with endometrial cancer versus controls. Increased interleukin 12 and decreased interleukin 10 secretion by blood monocytes in patients with endometrial cancer were also observed. CONCLUSIONS These findings showed that peripheral blood monocytes from patients with endometrial cancer show an altered phenotype and cytokine secretion when compared with controls. Limitations to this study include the small sample size, the need to investigate the effect of phenotype and cytokine changes in functional assays, as well as future studies investigating the effect on tumor-associated macrophages from endometrial tissue from cancer versus control patients. Nevertheless, these findings suggest that peripheral blood monocyte induced immunosuppression in endometrial cancer and implications in the design of future immunotargeting therapies remain to be elucidated.
Collapse
|
13
|
Khalili JS, Liu S, Rodríguez-Cruz TG, Whittington M, Wardell S, Liu C, Zhang M, Cooper ZA, Frederick DT, Li Y, Zhang M, Joseph RW, Bernatchez C, Ekmekcioglu S, Grimm E, Radvanyi LG, Davis RE, Davies MA, Wargo JA, Hwu P, Lizée G. Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 in melanoma. Clin Cancer Res 2012; 18:5329-40. [PMID: 22850568 DOI: 10.1158/1078-0432.ccr-12-1632] [Citation(s) in RCA: 251] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE In this study, we assessed the specific role of BRAF(V600E) signaling in modulating the expression of immune regulatory genes in melanoma, in addition to analyzing downstream induction of immune suppression by primary human melanoma tumor-associated fibroblasts (TAF). EXPERIMENTAL DESIGN Primary human melanocytes and melanoma cell lines were transduced to express WT or V600E forms of BRAF, followed by gene expression analysis. The BRAF(V600E) inhibitor vemurafenib was used to confirm targets in BRAF(V600E)-positive melanoma cell lines and in tumors from melanoma patients undergoing inhibitor treatment. TAF lines generated from melanoma patient biopsies were tested for their ability to inhibit the function of tumor antigen-specific T cells, before and following treatment with BRAF(V600E)-upregulated immune modulators. Transcriptional analysis of treated TAFs was conducted to identify potential mediators of T-cell suppression. RESULTS Expression of BRAF(V600E) induced transcription of interleukin 1 alpha (IL-1α) and IL-1β in melanocytes and melanoma cell lines. Further, vemurafenib reduced the expression of IL-1 protein in melanoma cell lines and most notably in human tumor biopsies from 11 of 12 melanoma patients undergoing inhibitor treatment. Treatment of melanoma-patient-derived TAFs with IL-1α/β significantly enhanced their ability to suppress the proliferation and function of melanoma-specific cytotoxic T cells, and this inhibition was partially attributable to upregulation by IL-1 of COX-2 and the PD-1 ligands PD-L1 and PD-L2 in TAFs. CONCLUSIONS This study reveals a novel mechanism of immune suppression sensitive to BRAF(V600E) inhibition, and indicates that clinical blockade of IL-1 may benefit patients with BRAF wild-type tumors and potentially synergize with immunotherapeutic interventions.
Collapse
Affiliation(s)
- Jahan S Khalili
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Joseph RW, Peddareddigari VR, Liu P, Miller PW, Overwijk WW, Bekele NB, Ross MI, Lee JE, Gershenwald JE, Lucci A, Prieto VG, McMannis JD, Papadopoulos N, Kim K, Homsi J, Bedikian A, Hwu WJ, Hwu P, Radvanyi LG. Impact of clinical and pathologic features on tumor-infiltrating lymphocyte expansion from surgically excised melanoma metastases for adoptive T-cell therapy. Clin Cancer Res 2011; 17:4882-91. [PMID: 21632855 DOI: 10.1158/1078-0432.ccr-10-2769] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE Clinical trials on adoptive T-cell therapy (ACT) using expanded tumor-infiltrating lymphocytes (TIL) have shown response rates of over 50% in refractory melanoma. However, little is known how clinical and pathologic features impact TIL outgrowth isolated from metastatic melanoma tumors. EXPERIMENTAL DESIGN We analyzed the impact of clinical and pathologic features on initial TIL outgrowth in 226 consecutive patients undergoing tumor resection. Successful initial TIL outgrowth was defined as ≥40 million viable lymphocytes harvested from all tumor fragments in a 5-week culture. To normalize for the different size of resected tumors and thus available tumor fragments, we divided the number of expanded TIL by the starting number of tumor fragments (TIL/fragment). RESULTS Overall, initial TIL outgrowth was successful in 62% of patients, with patients ≤30 years of age (94%; P = 0.01) and female patients (71% vs. 57% for males; P = 0.04) having the highest rate of success. Systemic therapy 30 days before tumor harvest negatively impacted initial TIL outgrowth compared to patients who never received systemic therapy (47% vs. 71%, P = 0.02). Biochemotherapy within 0 to 60 days of tumor harvest negatively impacted the initial TIL outgrowth with a success rate of only 16% (P < 0.0001). CONCLUSION Parameters such as age, sex, and the type and timing of prior systemic therapy significantly affect the success rate of the initial TIL outgrowth from tumor fragments for ACT; these parameters may be helpful in selecting patients for melanoma ACT.
Collapse
Affiliation(s)
- Richard W Joseph
- Department of Melanoma Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Ribas A, Weber JS, Chmielowski B, Comin-Anduix B, Lu D, Douek M, Ragavendra N, Raman S, Seja E, Rosario D, Miles S, Diamond DC, Qiu Z, Obrocea M, Bot A. Intra–Lymph Node Prime-Boost Vaccination against Melan A and Tyrosinase for the Treatment of Metastatic Melanoma: Results of a Phase 1 Clinical Trial. Clin Cancer Res 2011; 17:2987-96. [DOI: 10.1158/1078-0432.ccr-10-3272] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
16
|
Liu Y, He J, Xie X, Su G, Teitz-Tennenbaum S, Sabel MS, Lubman DM. Serum autoantibody profiling using a natural glycoprotein microarray for the prognosis of early melanoma. J Proteome Res 2010; 9:6044-51. [PMID: 20879797 DOI: 10.1021/pr100856k] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The poor prognosis of melanoma and the high cost of lymph node biopsy for melanoma patients have led to an urgent need for the discovery of convenient and accurate prognostic indicators. Here, we have developed a natural glycoprotein microarray to discover serum autoantibodies to distinguish between patients with node negative melanoma and node positive melanoma. Dual-lectin affinity chromatography was used to extract glycoproteins from a melanoma cell line. Liquid-based reverse phase separation and microarray platforms were then applied to separate and spot these natural proteins on nitrocellulose slides. The serum autoantibodies were investigated by exposing these proteins to sera from 43 patients that have already been diagnosed to have different stages of early melanoma. The combination of 9 fractions provides a 55% sensitivity with 100% specificity for the detection of node positive against node negative and a 62% sensitivity with 100% specificity for the detection of node negative against node positive. Recombinant proteins were used to confirm the results using a sample set with 79 patients with diagnosed melanoma. The response of sera against recombinant 94 kD glucose-regulated protein (GRP94), acid ceramidase (ASAH1), cathepsin D (CTSD), and lactate dehydrogenase B (LDHB) shared a similar pattern to the fractions where they were identified. The glycoarray platform provides a convenient and highly reproducible method to profile autoantibodies that could be used as serum biomarkers for prognosis of melanoma.
Collapse
Affiliation(s)
- Yashu Liu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Ma J, Usui Y, Takeuchi M, Okunuki Y, Kezuka T, Zhang L, Mizota A, Goto H. Human uveal melanoma cells inhibit the immunostimulatory function of dendritic cells. Exp Eye Res 2010; 91:491-9. [DOI: 10.1016/j.exer.2010.06.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 06/25/2010] [Accepted: 06/29/2010] [Indexed: 12/11/2022]
|
18
|
Hino R, Kabashima K, Kato Y, Yagi H, Nakamura M, Honjo T, Okazaki T, Tokura Y. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 2010; 116:1757-66. [PMID: 20143437 DOI: 10.1002/cncr.24899] [Citation(s) in RCA: 508] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND : Melanoma tends to be refractory to various immunotherapies because of tumor-induced immunosuppression. To investigate the mechanism underlining the immunosuppression of melanoma patients, the authors focused on programmed cell death-1 (PD-1)/PD-1 ligand 1 (PD-L1) interaction between tumor cells and T cells. METHODS : Melanoma specimens were collected from 59 primary tumors, 16 lymph nodes, and 4 lesions of in-transit metastasis. Specimens stained with anti-PD-L1 monoclonal antibodies were digitalized to jpg files. To evaluate the intensity of PD-L1 expression, histograms were used, and the red density (RD) was measured. PD-1 expression on T cells was analyzed in blood samples from 10 patients who had stage IV melanoma and in 4 samples of in-transit metastases. RESULTS : Twenty-five patients comprised the "low" PD-L1 expression group (RD value, <90), and 34 patients comprised the "high" group (RD value, > or =90). Breslow tumor thickness in the high-expression group was significantly higher than in the low-expression group. Univariate and multivariate analyses revealed that the overall survival rate of the high-expression group was significantly lower than that of the low-expression group. In all patients with stage IV disease who were examined, both CD8-positive and CD4-positive T cells had significantly higher PD-1 expression levels in the peripheral blood. Tumor-infiltrating T cells expressed high levels of PD-1, and its expression was elevated further during the clinical course. CONCLUSIONS : The current results indicated that there is a correlation between the degree of PD-L1 expression and the vertical growth of primary tumors in melanoma. Multivariate analysis demonstrated that PD-L1 expression is an independent prognostic factor for melanoma. Cancer 2010. (c) 2010 American Cancer Society.
Collapse
Affiliation(s)
- Ryosuke Hino
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Endometrial cancer is the most frequent gynecological cancer and the fourth most common cancer in women in the developed world. Over the last decade, immunotherapy has been the focus of intense investigation as a form of cancer treatment whereby the treatment initiates a host immune response ultimately eradicating the tumor. It has been suggested that in endometrial cancer and many other forms of cancer, immunosuppression poses a significant obstacle at inducing antitumor immunity by immunotherapy. This review will look at the different studies that have identified immunomodulation of T cells, cytokines and macrophages, and regulation of apoptotic and angiogenic factors in endometrial cancer patients that may contribute to the inefficiency of immunotherapy.
Collapse
Affiliation(s)
- Nicole Brooks
- School of Medical Sciences, RMIT University, Bundoora, Victoria, Australia
| | | |
Collapse
|
20
|
Abstract
Recent scientific advances have expanded our understanding of the immune system and its response to malignant cells. The clinical goal of tumour immunotherapy is to provide either passive or active immunity against malignancies by harnessing the immune system to target tumours. Monoclonal antibodies, cytokines, cellular immunotherapy, and vaccines have increasingly become successful therapeutic agents for the treatment of solid and haematological cancers in preclinical models, clinical trials, and practice. In this article, we review recent advances in the immunotherapy of cancer, focusing on new strategies and future perspectives as well as on clinical trials attempting to enhance the efficacy of immunotherapeutic modalities and translate this knowledge into effective cancer therapies.
Collapse
|
21
|
Fujita M, High WA, Asgari S, Lewis KD, Gonzalez R. Development of vitiligo during melanoma treatment with a novel survivin inhibitor: a case report and review of the literature. Int J Dermatol 2009; 48:426-30. [PMID: 19335434 DOI: 10.1111/j.1365-4632.2009.03987.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The development of vitiligo has been associated with an improved clinical response in melanoma patients. METHODS We report a case of vitiligo associated with a novel antisurvivin drug and review the literature to determine the pathogenesis of vitiligo occurring during melanoma treatment. RESULTS A 78-year-old man with stage IV malignant melanoma developed vitiligo after the first therapeutic cycle of a novel antisurvivin drug. Although his vitiligo remained static, his melanoma continued to progress and he died in 8 months. A review of the literature demonstrates a relationship between vitiligo development and improved clinical response in many melanoma cases treated with immunotherapy; however, the relationship may depend on the type of treatment. CONCLUSIONS Understanding complex immune responses in vitiliginous skin and melanoma sites is important in order to interpret the development of vitiligo occurring during melanoma treatment.
Collapse
Affiliation(s)
- Mayumi Fujita
- Department of Dermatology and Division of Medical Oncology, University of Colorado at Denver and Health Sciences Center, Aurora, Colorado 80010-7163, USA.
| | | | | | | | | |
Collapse
|
22
|
Mitchell DA, Sampson JH. Toward effective immunotherapy for the treatment of malignant brain tumors. Neurotherapeutics 2009; 6:527-38. [PMID: 19560742 PMCID: PMC2763142 DOI: 10.1016/j.nurt.2009.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 04/08/2009] [Accepted: 04/09/2009] [Indexed: 12/26/2022] Open
Abstract
The immunologic treatment of cancer has long been heralded as a targeted molecular therapeutic with the promise of eradicating tumor cells with minimal damage to surrounding normal tissues. However, a demonstrative example of the efficacy of immunotherapy in modulating cancer progression is still lacking for most human cancers. Recent breakthroughs in our understanding of the mechanisms leading to full T-cell activation, and recognition of the importance of overcoming tumor-induced immunosuppressive mechanisms, have shed new light on how to generate effective anti-tumor immune responses in humans, and sparked a renewed and enthusiastic effort to realize the full potential of cancer immunotherapy. The immunologic treatment of invasive malignant brain tumors has not escaped this re-invigorated endeavor, and promising therapies are currently under active investigation in dozens of clinical trials at several institutions worldwide. This review will focus on some of the most important breakthroughs in our understanding of how to generate potent anti-tumor immune responses, and some of the clear challenges that lie ahead in achieving effective immunotherapy for the majority of patients with malignant brain tumors. A review of immunotherapeutic strategies currently under clinical evaluation, as well as an outline of promising novel approaches on the horizon, is included to provide perspective on the active and stalwart progress toward effective immunotherapy for the treatment of malignant brain tumors.
Collapse
Affiliation(s)
- Duane A Mitchell
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA.
| | | |
Collapse
|
23
|
Haneke E. [Precancerous and early invasive carcinomas: non-surgical treatment of head and facial skin]. HNO 2009; 57:315-23. [PMID: 19322549 DOI: 10.1007/s00106-009-1892-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Chronic exposure to sunlight with its high proportion of high energy ultraviolet light is the main cause of the common cutaneous precancerous lesions and carcinomas of the head and neck. This causes a field cancerization effect frequently with multiple actinic keratoses (AKs), basal cell carcinomas (BCCs) and squamous cell carcinomas (SCCs). Although demonstrating the best cure rates and lowest recurrence rates, surgical excision rapidly progresses to its limits. Field cancerization requires field therapy. Non-specific caustic agents may remove superficial lesions, however, the modern therapeutic modalities such as topical cytotoxic treatment with 5-fluorouracil, photodynamic therapy with 5-aminolevulinic acid (ALA) or its methyl ester (MeALA), the topical immune response modifier imiquimod or the local application of the cyclooxygenase inhibitor diclofenac are more specific and effective. Intralesional and perilesional injections of cytotoxic agents and interferons as well as the new targeted anti-cancer drugs are further alternatives. The most important aspect, however, is the prophylaxis of chronic photodamage.
Collapse
Affiliation(s)
- E Haneke
- Dermatologische Klinik Inselspital, Univ Bern, Schweiz.
| |
Collapse
|
24
|
Mazorra Z, Mesa C, Fernández A, Fernández LE. Immunization with a GM3 ganglioside nanoparticulated vaccine confers an effector CD8(+) T cells-mediated protection against melanoma B16 challenge. Cancer Immunol Immunother 2008; 57:1771-80. [PMID: 18351335 PMCID: PMC11029898 DOI: 10.1007/s00262-008-0503-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Accepted: 03/03/2008] [Indexed: 01/11/2023]
Abstract
Preventive immunotherapy is an attractive strategy for patients at a high risk of having cancer. The success of prophylactic cancer vaccines would depend on the selection of target antigens that are essential for tumour growth and progression. The overexpression of GM3 ganglioside in murine and human melanomas and its important role in tumour progression makes this self antigen a potential target for preventive immunotherapy of this neoplasm. We have previously shown that preventive administration of a GM3-based vaccine to C57BL/6 mice elicited the rejection of the GM3 positive-B16 melanoma cells in most of the animals. Despite the crucial role of cellular immune response in tumour protection, the involvement of T cells in anti-tumour immunity of ganglioside vaccines is not described. Here, we examined the mechanisms by which this immunogen confers tumour protection. We have found that induction of anti-GM3 IgG antibodies correlated with tumour protection. Surprisingly, CD8(+) T cells, but not NK1.1(+) cells, are required in the effector phase of the antitumour immune response. The depletion of CD4(+) T cells during immunization phase did not affect the anti-tumour activity. In addition, T cells from surviving-immunized animals secreted IFNgamma when were co-cultured with IFNalpha-treated B16 melanoma cells or DCs pulsed with melanoma extract. Paradoxically, in spite of the glycolipidic nature of this antigen, these findings demonstrate the direct involvement of the cellular immune response in the anti-tumour protection induced by a ganglioside-based vaccine.
Collapse
Affiliation(s)
- Zaima Mazorra
- Department of Vaccines, Center of Molecular Immunology, Havana, Cuba.
| | | | | | | |
Collapse
|
25
|
Jackson AM, Mulcahy LA, Zhu XW, O'Donnell D, Patel PM. Tumour-mediated disruption of dendritic cell function: Inhibiting the MEK1/2-p44/42 axis restores IL-12 production and Th1-generation. Int J Cancer 2008; 123:623-32. [DOI: 10.1002/ijc.23530] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
26
|
Sarff M, Edwards D, Dhungel B, Wegmann KW, Corless C, Weinberg AD, Vetto JT. OX40 (CD134) expression in sentinel lymph nodes correlates with prognostic features of primary melanomas. Am J Surg 2008; 195:621-5; discussion 625. [PMID: 18374895 DOI: 10.1016/j.amjsurg.2007.12.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Revised: 12/20/2007] [Accepted: 12/20/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND The expression of OX40 (CD134) on activated CD4+ T cells has been associated with favorable cancer patient outcomes. Because of recent reports that sentinel lymph nodes (SLNs) may represent an immunosuppressive environment, we investigated the expression of OX40 in SLNs from patients with primary cutaneous melanoma. METHODS Samples of peripheral blood lymphocytes and a section of 71 SLNs from 53 patients with clinically node negative melanoma were purified for CD4+ T cells, stained for OX40, and analyzed by flow cytometry. RESULTS The mean percentage of OX40 on CD4 T cells in the SLNs versus peripheral blood lymphocytes was related indirectly to the T stage of the primary tumor and was decreased in ulcerated primary tumors and positive sentinel nodes. CONCLUSIONS The expression of OX40 on CD4+ T cells in SLNs draining primary melanomas decreased with more advanced tumor features (higher T stage, ulceration) and nodal involvement, suggesting that such tumors may have an immunosuppressive effect on the SLN microenvironment.
Collapse
Affiliation(s)
- MaryClare Sarff
- Earl A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Gamma-IFN-inducible-lysosomal thiol reductase modulates acidic proteases and HLA class II antigen processing in melanoma. Cancer Immunol Immunother 2008; 57:1461-70. [PMID: 18343923 DOI: 10.1007/s00262-008-0483-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Accepted: 02/08/2008] [Indexed: 12/19/2022]
Abstract
HLA class II-restricted antigen (Ag) processing and presentation are important for the activation of CD4+ T cells, which are the central orchestrating cells of immune responses. The majority of melanoma cells either expresses, or can be induced to express, HLA class II proteins. Thus, they are prime targets for immune mediated elimination by class II-restricted CD4+ T cells. We have previously shown that human melanoma cells lack an important enzyme, gamma interferon-inducible lysosomal thiol-reductase (GILT), capable of perturbing immune recognition of these tumors. Here, we show that GILT expression in human melanoma cells enhances Ag processing and presentation via HLA class II molecules. We also show that GILT expression influences the generation of active forms of cysteinyl proteases, cathepsins B, L and S, as well as an aspartyl protease cathepsin D in melanoma cells. Mechanistic studies revealed that GILT does not regulate acidic cathepsins at the transcriptional level; rather it colocalizes with the cathepsins and influences HLA class II Ag processing. GILT expression in melanoma cells also elevated HLA-DM molecules, which favor epitope loading onto class II in the endolysosomal compartments, enhancing CD4+ T cell recognition. These data suggest that GILT-expressing melanoma cells could prove to be very promising for direct antigen presentation and CD4+ T cell recognition, and may have direct implications for the design of cancer vaccines.
Collapse
|
28
|
Lizée G, Cantu MA, Hwu P. Less yin, more yang: confronting the barriers to cancer immunotherapy. Clin Cancer Res 2007; 13:5250-5. [PMID: 17875752 DOI: 10.1158/1078-0432.ccr-07-1722] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clinical trials involving T cell-based immunotherapy for the treatment of human cancer have shown limited degrees of success. In cancer vaccine trials conducted at multiple centers worldwide, immunization has often resulted in the robust elicitation of T cells that specifically recognize antigens expressed on the surface of tumor cells. However, to date, objective clinical responses resulting from these approaches have remained relatively rare. By contrast, adoptive transfer of laboratory-expanded T cells into patients has had more success, producing impressive clinical regressions in a subset of advanced metastatic melanoma patients. The failure of activated T cells to consistently induce clinical responses in many other patients has pushed us toward a deeper understanding of natural immunoregulatory mechanisms that are directly responsible for diminishing tumor-specific T-cell activation, migration, and effector function in vivo. Such immunosuppressive factors likely evolved to prevent autoimmunity, but are frequently co-opted by tumors to evade tumor-specific immune responses. With this knowledge, it now becomes imperative to develop specific clinical interventions capable of eliminating tumor-specific immunosuppression, with the goal of shifting the balance to favor effector T-cell function and tumor cell killing.
Collapse
Affiliation(s)
- Gregory Lizée
- Department of Melanoma Medical Oncology, M. D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
29
|
Paulos CM, Kaiser A, Wrzesinski C, Hinrichs CS, Cassard L, Boni A, Muranski P, Sanchez-Perez L, Palmer DC, Yu Z, Antony PA, Gattinoni L, Rosenberg SA, Restifo NP. Toll-like receptors in tumor immunotherapy. Clin Cancer Res 2007; 13:5280-9. [PMID: 17875756 PMCID: PMC2131730 DOI: 10.1158/1078-0432.ccr-07-1378] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lymphodepletion with chemotherapeutic agents or total body irradiation (TBI) before adoptive transfer of tumor-specific T cells is a critical advancement in the treatment of patients with melanoma. More than 50% of patients that are refractory to other treatments experience an objective or curative response with this approach. Emerging data indicate that the key mechanisms underlying how TBI augments the functions of adoptively transferred T cells include (a) the depletion of regulatory T cells (T(reg)) and myeloid-derived suppressor cells that limit the function and proliferation of adoptively transferred cells; (b) the removal of immune cells that act as "sinks" for homeostatic cytokines, whose levels increase after lymphodepletion; and (c) the activation of the innate immune system via Toll-like receptor 4 signaling, which is engaged by microbial lipopolysaccharide that translocated across the radiation-injured gut. Here, we review these mechanisms and focus on the effect of Toll-like receptor agonists in adoptive immunotherapy. We also discuss alternate regimens to chemotherapy or TBI, which might be used to safely treat patients with advanced disease and promote tumor regression.
Collapse
Affiliation(s)
- Chrystal M Paulos
- National Cancer Institute, NIH, Mark O. Hatfield Clinical Research Center, Bethesda, Maryland 20892-1502, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
An often overlooked facet of tumor biology research is the involvement of the surrounding tumor microenvironment. Increasing evidence is being presented to support a major role for stromal components in all stages of tumorigenesis including initiation, progression, and metastasis. Melanoma serves as a model for studying cellular and stromal interactions within the tumor microenvironment due to the array of cell types localized to these lesions. Here, we discuss the both the molecular mechanisms, as well as the extracellular and contextual input that contribute to melanoma progression. Special emphasis is given to the assorted cell types and their interactions with the extracellular matrix and adjacent cells. Melanoma progression also initiates development of intralesional hypoxic regions; the relative significance of hypoxia in disease is also addressed. Lastly, a number of laboratories are currently developing innovative strategies to study melanoma within a microenvironmental platform. These promising model systems and their potential for closing current gaps in knowledge of disease are reviewed. The development of such models holds translational value that cannot be achieved with most current systems.
Collapse
Affiliation(s)
- John T Lee
- The Wistar Institute, Program of Molecular and Cellular Oncogenesis, Philadelphia, PA 19104, USA
| | | |
Collapse
|
31
|
Ahmadzadeh M, Antony PA, Rosenberg SA. IL-2 and IL-15 each mediate de novo induction of FOXP3 expression in human tumor antigen-specific CD8 T cells. J Immunother 2007; 30:294-302. [PMID: 17414320 PMCID: PMC2174606 DOI: 10.1097/cji.0b013e3180336787] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although FOXP3 is primarily expressed by regulatory CD4 T cells (Treg) in vivo, polyclonal activation of human CD8 T cells can result in the expression of FOXP3 in a fraction of CD8 T cells. However, the cellular lineage and mechanism of FOXP3 induction in CD8 T cells remain unclear. Here, we demonstrate that interleukin-2 (IL-2) induces FOXP3 expression in OKT3-stimulated or antigen-stimulated CD8 T cells, indicating that FOXP3 expression is neither limited to a unique subset of CD8 T cells nor dependent on the mode of T-cell receptor stimulation. In the absence of IL-2, antigen stimulation resulted in T-cell activation and acquisition of effector function without induction of FOXP3, indicating that acquisition of effector function is independent of induction of FOXP3 expression in CD8 T cells. Interestingly, IL-15, but not IL-7 or IL-21, also led to de novo induction of FOXP3 in antigen-specific CD8 T cells, suggesting that signaling by IL-2/IL-15Rbeta chain is pivotal for induction of FOXP3 in human CD8 T cells. These findings indicate that induction of FOXP3 is intrinsic to CD8 T cells that are activated in the presence of IL-2 or IL-15, and in vitro-induced expression of FOXP3 cannot be simply interpreted as an indicator of Treg activity or activation marker.
Collapse
Affiliation(s)
- Mojgan Ahmadzadeh
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
32
|
Celis E. Overlapping human leukocyte antigen class I/II binding peptide vaccine for the treatment of patients with stage IV melanoma. Cancer 2007; 110:203-14. [PMID: 17541944 DOI: 10.1002/cncr.22744] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND MPS160 (GRAMLGTHTMEVTV) is a glycoprotein 100-derived melanoma peptide that contains overlapping human leukemic antigen A2-, DR53-, and DQw6-restricted T-cell epitopes. In preclinical testing, MPS160 demonstrated superior immunization and antitumor activity. In this report, the authors present the results from a clinical trial that evaluated the safety and immunologic efficacy of the MPS160 vaccine in patients with metastatic melanoma. METHODS Patients with stage IV melanoma were randomized to 1 of 3 treatment arms: 1) MPS160 in incomplete Freund adjuvant (Montanide ISA-51); 2) MPS160 in Montanide ISA-51 with 75 microg of granulocyte-macrophage-stimulating factor (GM-CSF); or 3) MPS160 in Montanide ISA-51 with 100 microg of GM-CSF. Vaccines were administered every 3 weeks until patients developed disease progression or severe toxicity. Patients were aged >or=18 years with metastatic melanoma and a good performance status. Exclusion criteria included pregnancy/nursing, brain metastases, and ongoing chemotherapy. Immunologic efficacy was ascertained by using tetramer and functional analysis of peripheral blood lymphocytes. RESULTS None of the 28 patients exhibited objective tumor responses or severe toxicities. Four patients remained progression free for >or=100 days. Immunologic analysis was available for 21 patients. Laboratory data demonstrated 1) increased frequency of vaccine-specific, nonfunctional cytotoxic T lymphocytes in 10 patients; 2) no differences in immunization efficacy among the treatment arms; and 3) evidence of systemic cytokine/immune dysfunction. CONCLUSIONS Clinically, the MPS160 vaccine was ineffective. Phenotypic (tetramer) evidence of immunization was ineffective functionally and most likely was caused by global immune dysfunction, as illustrated by abnormal cytokine profiles in peripheral blood. In this report, the authors discuss possible implications of the current results on future cancer vaccine studies.
Collapse
|