1
|
Schatten H. Immunodiagnostics and Immunotherapy Possibilities for Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1096:185-194. [PMID: 30324354 DOI: 10.1007/978-3-319-99286-0_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite significant progress in early detection and improved treatment modalities prostate cancer remains the second leading cause of cancer death in American men which results in about 30,000 deaths per year in the USA. An aggressive phenotype leading to 2.58% risk of dying from prostate cancer still exists and immunotherapy has offered new possibilities to treat metastatic prostate cancer that cannot be treated by other modalities. Cancer immunotherapy is a rapidly growing field of research aimed at identifying biomarkers in immunodiagnosis and to develop new therapies by enabling the immune system to detect and destroy cancer cells. Immunotherapy falls into three different broad categories which are checkpoint inhibitors, cytokines, and vaccine immunotherapy. While immunotherapy to treat prostate cancer is still limited progress has been made; for treatment of advanced prostate cancer sipuleucel-T has been administered to patients in personalized doses to destroy prostate cancer cells which is promising and invites further research to determine immunotherapies for advanced prostate cancer. Antibody-based targeted immunotherapy and dendritic-cell-based vaccination are among the therapies that are currently being evaluated as promising approaches to treat prostate cancer. Combination immunotherapies include prostate cancer vaccines and radiotherapy for castration resistant prostate cancer. Microbial vectors for prostate cancer immunotherapy have been developed and bacterial strains have been engineered to express cancer-specific antigens, cytokines, and prodrug-converting cytokines. These approaches are addressed in the present review.
Collapse
Affiliation(s)
- Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
2
|
Pham T, Carpinteri S, Sampurno S, Pereira L, Roth S, Narasimhan V, Darcy P, Desai J, Heriot AG, Ramsay RG. Novel Vaccine Targeting Colonic Adenoma: a Pre-clinical Model. J Gastrointest Surg 2019; 23:626-633. [PMID: 30623377 DOI: 10.1007/s11605-018-4060-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/15/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is the second leading cause of cancer-related mortality in the USA. Over 80% of CRC develop from adenomatous polyps. Hence, early treatment and prevention of adenomas would lead to a significant decrease of disease burden for CRC. MYB is a transcription factor that is overexpressed in both precancerous adenomatous polyps and colorectal cancer, and hence an ideal immunotherapeutic target. We have developed a cancer vaccine, TetMYB, that targets MYB and aim to evaluate its efficacy in the prophylactic and therapeutic management of adenomatous polyps. MATERIAL AND METHODS Six- to eight-week-old Apcmin/+ (Familial Adenomatous Polyposis model) and Apc580S (sporadic model) C57BL/6 mice were used. The Apcmin/+ mice are carried a germline mutation of one Apc allele whereas the Apc580S model has an inducible silencing of one Apc allele, when exposed to tamoxifen, via the Cre-Lox recombination enzyme system. In the prophylactic treatment group, Apcmin/+ and Apc580S C57BL/6 mice were vaccinated and surveyed for clinical signs of distress. Number of adenoma and survival were measured. In the therapeutic cohort, Apc580S C57BL/6 mice were given tamoxifen-laced food to activate Cre-Lox recombinase mediated silencing of one Apc allele and thus inducing adenoma development. Following adenoma detection, mice were vaccinated with TetMYB and treated with anti-PD-1 antibody and were analyzed for overall survival. RESULTS In both the prophylactic and therapeutic setting, mice vaccinated with TetMYB had a significantly improved outcome, with the vaccinated Apcmin/+ mice having a median survival benefit of 70 days (p = 0.008) and the vaccinated Apc580S mice having a mean survival benefit of 134 days (p = 0.01) over the unvaccinated mice. In the prophylactic cohort, immunofluorescence confirmed a stronger cytotoxic CD8+ T cell infiltrate in the vaccinated group, implying an anti-tumor immune response. In the therapeutic cohort, vaccinated Apc580S mice showed significantly reduced adenoma progression rate compared to the unvaccinated mice (p = 0.0005). CONCLUSION TetMYB vaccine has shown benefit in a prophylactic and therapeutic setting in the management of colonic adenoma in a murine model. This will form the basis for a future clinical trial to prevent and treat colonic adenomatous polyps.
Collapse
Affiliation(s)
- Toan Pham
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia. .,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia. .,Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Sandra Carpinteri
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Shienny Sampurno
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Lloyd Pereira
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Sara Roth
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Vignesh Narasimhan
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Phillip Darcy
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Jayesh Desai
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alexander G Heriot
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Robert G Ramsay
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia.,Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Pham T, Roth S, Kong J, Guerra G, Narasimhan V, Pereira L, Desai J, Heriot A, Ramsay R. An Update on Immunotherapy for Solid Tumors: A Review. Ann Surg Oncol 2018; 25:3404-3412. [DOI: 10.1245/s10434-018-6658-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Indexed: 12/29/2022]
|
4
|
Overgaard NH, Principe DR, Schachtschneider KM, Jakobsen JT, Rund LA, Grippo PJ, Schook LB, Jungersen G. Genetically Induced Tumors in the Oncopig Model Invoke an Antitumor Immune Response Dominated by Cytotoxic CD8β + T Cells and Differentiated γδ T Cells Alongside a Regulatory Response Mediated by FOXP3 + T Cells and Immunoregulatory Molecules. Front Immunol 2018; 9:1301. [PMID: 29930558 PMCID: PMC5999797 DOI: 10.3389/fimmu.2018.01301] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/25/2018] [Indexed: 12/31/2022] Open
Abstract
In recent years, immunotherapy has shown considerable promise in the management of several malignancies. However, the majority of preclinical studies have been conducted in rodents, the results of which often translate poorly to patients given the substantial differences between murine and human immunology. As the porcine immune system is far more analogous to that of humans, pigs may serve as a supplementary preclinical model for future testing of such therapies. We have generated the genetically modified Oncopig with inducible tumor formation resulting from concomitant KRASG12D and TP53R167H mutations under control of an adenoviral vector Cre-recombinase (AdCre). The objective of this study was to characterize the tumor microenvironment in this novel animal model with respect to T-cell responses in particular and to elucidate the potential use of Oncopigs for future preclinical testing of cancer immunotherapies. In this study, we observed pronounced intratumoral T-cell infiltration with a strong CD8β+ predominance alongside a representation of highly differentiated γδ T cells. The infiltrating CD8β+ T cells displayed increased expression of the cytotoxic marker perforin when compared with the peripheral T-cell pool. Similarly, there was robust granzyme B staining localizing to the tumors; affirming the presence of cytotoxic immune cells within the tumor. In parallel with this antitumor immune response, the tumors displayed enrichment in FOXP3-expressing T cells and increased gene expression of indoleamine 2,3-dioxygenase 1 (IDO1), cytotoxic T-lymphocyte-associated protein 4 (CTLA4), and programmed death-ligand 1 (PDL1). Finally, we investigated the Oncopig immune system in mediating antitumor immunity. We observed pronounced killing of autologous tumor cells, which demonstrates the propensity of the Oncopig immune system to recognize and mount a cytotoxic response against tumor cells. Together, these findings suggest innate and adaptive recognition of the induced tumors with a concomitant in vivo suppression of T-cell effector functions. Combined, the data support that the Oncopig may serve as a valuable model for future preclinical testing of immunotherapies aimed at reactivating tumor-directed cytotoxicity in vivo.
Collapse
Affiliation(s)
- Nana H Overgaard
- Department of Micro- and Nanotechnology, Technical University of Denmark, Kongens Lyngby, Denmark.,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, United States
| | | | - Jeanne Toft Jakobsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Laurie A Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Urbana-Champaign, Chicago, IL, United States
| | - Lawrence B Schook
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Department of Radiology, University of Illinois, Chicago, IL, United States
| | - Gregers Jungersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
5
|
Kakhi Z, Frisch B, Bourel-Bonnet L, Hemmerlé J, Pons F, Heurtault B. Airway administration of a highly versatile peptide-based liposomal construct for local and distant antitumoral vaccination. Int J Pharm 2015; 496:1047-56. [PMID: 26602294 DOI: 10.1016/j.ijpharm.2015.11.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/13/2015] [Accepted: 11/14/2015] [Indexed: 01/28/2023]
Abstract
With the discovery of tumor-associated antigens such as ErbB2, vaccination is considered as a promising strategy to prevent the development of cancer or treat the existing disease. Among routes of immunization, the respiratory route provides the opportunity to develop non-invasive approach for vaccine delivery. In the current study, this administration route was used in order to investigate the potency of a highly versatile di-epitopic liposomal construct to exhibit local or distant antitumoral efficiency after prophylactic or therapeutic vaccination in mice. Well-characterized liposomes, containing the ErbB2 (p63-71) TCD8(+) and HA (p307-319) TCD4(+) peptide epitopes and the Pam2CAG adjuvant, were formulated and administered into the airway of naïve BALB/c mice. The nanoparticle vaccine candidate induced local and specific systemic immune response, as measured by immune cell infiltration and chemokine and cytokine production in BALF or lung tissue, and by spleen T-cell activation ex vivo, respectively. This potent immune response resulted in an efficient antitumor activity against both lung and solid s.c. tumors. Interestingly, the antitumor efficacy was observed after both prophylactic and therapeutic vaccinations, which are the most judicious ones to fight cancer. Our data showed an undeniable interest of liposomal peptide-based vaccines in antitumor vaccination by the respiratory route, opening new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Zahra Kakhi
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Benoît Frisch
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Line Bourel-Bonnet
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | | | - Françoise Pons
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France.
| | - Béatrice Heurtault
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France.
| |
Collapse
|
6
|
Overgaard NH, Frøsig TM, Welner S, Rasmussen M, Ilsøe M, Sørensen MR, Andersen MH, Buus S, Jungersen G. Establishing the pig as a large animal model for vaccine development against human cancer. Front Genet 2015; 6:286. [PMID: 26442104 PMCID: PMC4584933 DOI: 10.3389/fgene.2015.00286] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/28/2015] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy has increased overall survival of metastatic cancer patients, and cancer antigens are promising vaccine targets. To fulfill the promise, appropriate tailoring of the vaccine formulations to mount in vivo cytotoxic T cell (CTL) responses toward co-delivered cancer antigens is essential. Previous development of therapeutic cancer vaccines has largely been based on studies in mice, and the majority of these candidate vaccines failed to induce therapeutic responses in the subsequent human clinical trials. Given that antigen dose and vaccine volume in pigs are translatable to humans and the porcine immunome is closer related to the human counterpart, we here introduce pigs as a supplementary large animal model for human cancer vaccine development. IDO and RhoC, both important in human cancer development and progression, were used as vaccine targets and 12 pigs were immunized with overlapping 20mer peptides spanning the entire porcine IDO and RhoC sequences formulated in CTL-inducing adjuvants: CAF09, CASAC, Montanide ISA 51 VG, or PBS. Taking advantage of recombinant swine MHC class I molecules (SLAs), the peptide-SLA complex stability was measured for 198 IDO- or RhoC-derived 9-11mer peptides predicted to bind to SLA-1*04:01, −1*07:02, −2*04:01, −2*05:02, and/or −3*04:01. This identified 89 stable (t½ ≥ 0.5 h) peptide-SLA complexes. By IFN-γ release in PBMC cultures we monitored the vaccine-induced peptide-specific CTL responses, and found responses to both IDO- and RhoC-derived peptides across all groups with no adjuvant being superior. These findings support the further use of pigs as a large animal model for vaccine development against human cancer.
Collapse
Affiliation(s)
- Nana H Overgaard
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Thomas M Frøsig
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Simon Welner
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Michael Rasmussen
- Department of International Health, Immunology and Microbiology, University of Copenhagen Copenhagen, Denmark
| | - Mette Ilsøe
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Maria R Sørensen
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Mads H Andersen
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Denmark
| | - Søren Buus
- Department of International Health, Immunology and Microbiology, University of Copenhagen Copenhagen, Denmark
| | - Gregers Jungersen
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| |
Collapse
|
7
|
|
8
|
Bot A, Marincola F, Smith KA. Repositioning therapeutic cancer vaccines in the dawning era of potent immune interventions. Expert Rev Vaccines 2013; 12:1219-34. [PMID: 24099049 DOI: 10.1586/14760584.2013.836908] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Based on lessons learned with various immune interventions, this review aims to provide a constructive framework for repositioning therapeutic cancer vaccination. Intensive research throughout the past decade has identified key hurdles interfering with the efficacy of cancer vaccines. The vaccination concept still holds promise if positioned appropriately in minimal residual disease and select early disease stage cancer indications. However, in advanced cancer, it must be integrated with complementary immune interventions to ensure reconstruction of a functional immune repertoire and simultaneous blockade of immune inhibiting mechanisms. Vaccination could render complex and integrative immune interventions simpler, safer and more effective. The near future will witness an explosion of activities in the cancer immunotherapy arena, witnessing a rational repositioning of vaccines rather than their extinction.
Collapse
|
9
|
Rodriguez PC, Sanchez B. Challenges and opportunities for cancer vaccines in the current NSCLC clinical scenario. Curr Top Med Chem 2013; 13:2551-61. [PMID: 24066886 PMCID: PMC4104452 DOI: 10.2174/15680266113136660182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/25/2013] [Accepted: 08/02/2013] [Indexed: 12/20/2022]
Abstract
This review is aimed to focus on NSCLC as an emerging and promising model for active immunotherapy and the challenges for its inclusion in the current clinical scenario. Cancer vaccines for NSCLC have been focused as a therapeutic option based on the identification of a tumor hallmark and the active immunization with the related molecules that triggers cellular and/or humoral responses that consequently destroy or delay the rate of malignant progression. This therapeutic intervention in an established disease state has been aimed to impact into prolonging patient´s survival with ethically accepted quality of life. Understanding of relationship between structure and function in cancer vaccines is essential to interpret their opportunities to impact into prolonging survival and increasing quality of life in cancer patients. It is widely accepted that the failure of the cancer vaccines in the NSCLC scenario is related with its introduction in the advanced disease stages and poor performance status of the patients due to the combination of the tumor induced immunosuppression with the immune senescence. Despite first, second and emerging third line of onco-specific treatments the life expectancy for NSCLC patients diagnosed at advanced stages is surrounding the 12 months of median survival and in facts the today real circumstances are extremely demanding for the success inclusion of cancer vaccines as therapeutic choice in the clinical scenario. The kinetics of the active immunizations encompasses a sequential cascade of clinical endpoints: starting by the activation of the immune system, followed by the antitumor response and finalizing with the consequential impact on patients’ overall survival. Today this cascade of clinical endpoints is the backbone for active immunization assessment and moreover the concept of cancer vaccines, applied in the NSCLC setting, is just evolving as a complex therapeutic strategy, in which the opportunities for cancer vaccines start from the selection of the target cancer hallmark, followed by the vaccine formulation and its platforms for immune potentiating, also cover the successful insertion in the standard of care, the chronic administration beyond progression disease, the personalization based on predictors of response and the potential combination with other targeted therapies.
Collapse
Affiliation(s)
- Pedro C Rodriguez
- Center of Molecular Immunology, Clinical Research Direction, 216 and 15, Playa, P.O.Box: 16040, Havana 11600, Cuba.
| | | |
Collapse
|
10
|
Abstract
Prostate cancer remains a significant health problem for men in the Western world. Although treatment modalities are available, these do not confer long-term benefit and are accompanied by deleterious side effects. Immunotherapy represents a valuable alternative to conventional treatments by inducing tumour-specific immune responses that control the growth of cancer cells. Sipuleucel-T is approved by the FDA as an immunotherapeutic agent for the treatment of patients with asymptomatic or minimally symptomatic castration-resistant prostate cancer (CRPC). Although this approval has raised cost-versus-benefit issues, it has provided proof of concept for the therapeutic potential of active immunotherapy approaches for metastatic CRPC. Numerous clinical studies have demonstrated clinical benefit using immunotherapy compared to traditional chemotherapy and several active immunotherapy approaches (at various developmental stages)have demonstrated the potential to change the face of prostate cancer treatment.
Collapse
|
11
|
Kuo CH, Lo CY, Chung FT, Lee KY, Lin SM, Wang CH, Heh CC, Chen HC, Kuo HP. Concomitant active tuberculosis prolongs survival in non-small cell lung cancer: a study in a tuberculosis-endemic country. PLoS One 2012; 7:e33226. [PMID: 22438899 PMCID: PMC3306389 DOI: 10.1371/journal.pone.0033226] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 02/09/2012] [Indexed: 01/13/2023] Open
Abstract
Background Adjuvant tumor cell vaccine with chemotherapy against non-small cell lung cancer (NSCLC) shows limited clinical response. Whether it provokes effective cellular immunity in tumor microenvironment is questionable. Concomitant active tuberculosis in NSCLC (TBLC) resembles locoregional immunotherapy of tumor cell vaccine; thus, maximally enriches effective anti-tumor immunity. This study compares the survival and immunological cell profile in TBLC over NSCLC alone. Methods Retrospective review of NSCLC patients within 1-year-period of 2007 and follow-up till 2010. Results A total 276 NSCLC patients were included. The median survival of TBLC is longer than those of NSCLC alone (11.6 vs. 8.8 month, p<0.01). Active tuberculosis is an independent predictor of better survival with HR of 0.68 (95% CI, 0.48∼0.97). Squamous cell carcinoma (SCC) (55.8 vs. 31.7%, p<0.01) is a significant risk factor for NSCLC with active TB. The median survival of SCC with active tuberculosis is significantly longer than adenocarcinoma or undetermined NSCLC with TB (14.2 vs. 6.6 and 2.8 months, p<0.05). Active tuberculosis in SCC increases the expression of CD3 (46.4±24.8 vs. 24.0±16.0, p<0.05), CXCR3 (35.1±16.4 vs. 19.2±13.3, p<0.01) and IP-10 (63.5±21.9 vs. 35.5±21.0, p<0.01), while expression of FOXP3 is decreased (3.5±0.5 vs. 13.3±3.7 p<0.05, p<0.05). Survival of SCC with high expression of CD3 (12.1 vs. 3.6 month, p<0.05) and CXCR3 (12.1 vs. 4.4 month, p<0.05) is longer than that with low expression. Conclusions Active tuberculosis in NSCLC shows better survival outcome. The effective T lymphocyte infiltration in tumor possibly underlies the mechanism. Locoregional immunotherapy of tumor cell vaccine may deserve further researches.
Collapse
Affiliation(s)
- Chih-Hsi Kuo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
- Department of Thoracic Medicine, St. Paul’s Hospital, Taoyuan, Taiwan
| | - Chun-Yu Lo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Fu-Tsai Chung
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Kang-Yun Lee
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chun-Hua Wang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chih-Chen Heh
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Hao-Cheng Chen
- Department of Thoracic Medicine, St. Paul’s Hospital, Taoyuan, Taiwan
- Department of Thoracic Medicine, Taipei Medical University, Shuang-Ho Hospital, Taipei, Taiwan
| | - Han-Pin Kuo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
12
|
Activation of anti-tumor immune response and reduction of regulatory T cells with Mycobacterium indicus pranii (MIP) therapy in tumor bearing mice. PLoS One 2011; 6:e25424. [PMID: 21984926 PMCID: PMC3184142 DOI: 10.1371/journal.pone.0025424] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 09/05/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Role of immune system in protecting the host from cancer is well established. Growing cancer however subverts immune response towards Th2 type and escape from antitumor mechanism of the host. Activation of both innate and Th1 type response is crucial for host antitumor activity. In our previous study it was found, that Mycobacterium indicus pranii (MIP) also known as M. w induces Th1 type response and activates macrophages in animal model of tuberculosis. Hence, we studied the immunotherapeutic potential of MIP in mouse tumor model and the underlying mechanisms for its antitumor activity. METHODOLOGY AND PRINCIPAL FINDINGS Tumors were implanted by injecting B16F10 melanoma cells subcutaneously into C57BL/6 mice. Using the optimized dose and treatment regimes, anti-tumor efficacy of heat killed MIP was evaluated. In MIP treated group, tumor appeared in only 50-60% of mice, tumor growth was delayed and tumor volume was less as compared to control. MIP mediated immune activation was analysed in the tumor microenvironment, tumor draining lymph node and spleen. Induction of Th1 response and higher infiltration of immune cells in the tumor microenvironment was observed in MIP treated mice. A large fraction of these immune cells were in activated state as confirmed by phenotypic and functional analysis. Interestingly, percentage of Treg cells in the tumor milieu of treated mice was less. We also evaluated efficacy of MIP along with chemotherapy and found a better response as compared to chemotherapy alone. CONCLUSION MIP therapy is effective in protecting mice from tumor. It activates the immune cells, increases their infiltration in tumor, and abrogates tumor mediated immune suppression.
Collapse
|
13
|
Sørensen RB, Junker N, Kirkin A, Voigt H, Svane IM, Becker JC, thor Straten P, Andersen MH. The immunodominant HLA-A2-restricted MART-1 epitope is not presented on the surface of many melanoma cell lines. Cancer Immunol Immunother 2009; 58:665-75. [PMID: 18828018 PMCID: PMC11030848 DOI: 10.1007/s00262-008-0588-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Accepted: 08/30/2008] [Indexed: 10/21/2022]
Abstract
Among the relatively large number of known tumor-associated antigens (TAA) which are recognized by human CD8 T-cells, Melan-A/MART-1 is one of the most-if not the most-frequently used target for anti-cancer vaccines in HLA-A2 + melanoma patients. In this study, we analyzed the killing of a large panel of melanoma cells by a high avidity, MART-1-specific T-cell clone or a MART-1-specific, polyclonal T-cell culture. Strikingly, we observed that the MART-1-specific T-cells only killed around half of the analyzed melanoma cell lines. In contrast a Bcl-2-specific T-cell clone killed all melanoma cell lines, although the T-cell avidity of this clone was significantly lower. The MART-1-specific T-cell clone expressed NKG-2D and was fully capable of releasing both perforin and Granzyme B. Notably, the resistance to killing by the MART-1-specific T-cells could be overcome by pulsing of the melanoma cells with the MART-1 epitope. Thus, the very frequently used MART-1 epitope was not expressed on the surface of many melanoma cell lines. Our data emphasize that the selected tumor antigens and/or epitopes are critical for the outcome of anti-cancer immunotherapy.
Collapse
MESH Headings
- Antigens, Neoplasm/analysis
- Antigens, Neoplasm/immunology
- Antigens, Surface/analysis
- Antigens, Surface/immunology
- Cell Line, Tumor/chemistry
- Cell Line, Tumor/immunology
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic
- Epitopes/analysis
- Epitopes/immunology
- Epitopes, T-Lymphocyte/analysis
- Epitopes, T-Lymphocyte/immunology
- Granzymes/biosynthesis
- Granzymes/immunology
- HLA-A2 Antigen/immunology
- Humans
- Immunodominant Epitopes/analysis
- Immunodominant Epitopes/immunology
- Interferon-gamma/metabolism
- Melanoma/chemistry
- Melanoma/immunology
- NK Cell Lectin-Like Receptor Subfamily K/biosynthesis
- NK Cell Lectin-Like Receptor Subfamily K/immunology
- Neoplasm Proteins/analysis
- Neoplasm Proteins/immunology
- Perforin
- Pore Forming Cytotoxic Proteins/biosynthesis
- Pore Forming Cytotoxic Proteins/immunology
- Proto-Oncogene Proteins c-bcl-2/immunology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Rikke Bæk Sørensen
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev University Hospital, 2730 Herlev, Denmark
| | - Niels Junker
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev University Hospital, 2730 Herlev, Denmark
| | - Alexei Kirkin
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev University Hospital, 2730 Herlev, Denmark
| | - Heike Voigt
- Department of Dermatology, University of Würzburg, 97080 Würzburg, Germany
| | - Inge Marie Svane
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev University Hospital, 2730 Herlev, Denmark
| | - Jürgen C. Becker
- Department of Dermatology, University of Würzburg, 97080 Würzburg, Germany
| | - Per thor Straten
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev University Hospital, 2730 Herlev, Denmark
| | - Mads Hald Andersen
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev University Hospital, 2730 Herlev, Denmark
| |
Collapse
|
14
|
Zhao L, Liu Z, Fan D. Overview of mimotopes and related strategies in tumor vaccine development. Expert Rev Vaccines 2009; 7:1547-55. [PMID: 19053210 DOI: 10.1586/14760584.7.10.1547] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tumor vaccine has been studied extensively as an alternative or adjuvant therapy in the treatment of malignant tumors in the hope of prolonging the overall survival rates of cancer patients. The efficacy largely relies on the specificity of the target. In the last decade, many antibody epitopes, called mimotopes, have been revealed as candidates through phage-display technology. These mimotopes do not necessarily consist of amino acid sequences that are identical to the native antigen but they do mimic their structure. Tumor vaccines based on these mimotopes have been proposed as an important developing strategy. Some peptide mimotopes have produced encouraging clinical outcomes. Although most studies are still in the preclinical phase, these findings will possibly pave the way for the development of novel mimotope-based tumor vaccines.
Collapse
Affiliation(s)
- Lina Zhao
- State Key Laboratory of Cancer Biology, Institute of Digestive Disease, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | | | | |
Collapse
|
15
|
Yagui-Beltrán A, Coussens LM, Jablons DM. Respiratory Homeostasis and Exploitation of the Immune System for Lung Cancer Vaccines. ACTA ACUST UNITED AC 2009; 58:40-48. [PMID: 22368692 DOI: 10.17925/ohr.2009.05.1.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Lung cancer is the leading cause of all cancer deaths in the US. The international scientific and clinical community has made significant advances toward understanding specific molecular mechanisms underlying lung carcinogenesis; however, despite these insights and advances in surgery and chemoradiotherapy, the prognosis for non-small-cell lung cancer (NSCLC) remains poor. Nonetheless, significant effort is being focused on advancing translational research evaluating the efficacy of novel targeted therapeutic strategies for lung cancer. Illustrative examples of this include antagonists of the epidermal growth factor receptor (EGFR), tyrosine kinase inhibitors (TKIs) such as gefitinib and erlotinib, and a diverse assortment of anti-angiogenic compounds targeting growth factors and/or their receptors that regulate tumor-associated angiogenic programs. In addition, with the increased awareness of the significant role chronically activated leukocytes play as potentiators of solid-tumor development, the role of innate and adaptive immune cells as regulators of lung carcinogenesis is being examined. While some of these studies are examining how novel therapeutic strategies may enhance the efficacy of lung cancer vaccines, others are evaluating the intrinsic characteristics of the immune response to lung cancer in order to identify rate-limiting molecular and/or cellular programs to target with novel anticancer therapeutics. In this article, we explore important aspects of the immune system and its role in regulating normal respiratory homeostasis compared with the immune response accompanying development of lung cancer. These hallmarks are then discussed in the context of recent efforts to develop lung cancer vaccines, where we have highlighted important concepts that must be taken into consideration for future development of novel therapeutic strategies and clinical trials assessing their efficacy.
Collapse
Affiliation(s)
- Adam Yagui-Beltrán
- Post-doctoral Fellow, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco
| | | | | |
Collapse
|
16
|
Wenandy L, Sørensen RB, Svane IM, thor Straten P, Andersen MH. RhoC a new target for therapeutic vaccination against metastatic cancer. Cancer Immunol Immunother 2008; 57:1871-8. [PMID: 18415097 PMCID: PMC11030672 DOI: 10.1007/s00262-008-0517-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Accepted: 03/29/2008] [Indexed: 10/22/2022]
Abstract
Most cancer deaths are due to the development of metastases. Increased expression of RhoC is linked to enhanced metastatic potential in multiple cancers. Consequently, the RhoC protein is an attractive target for drug design. The clinical application of immunotherapy against cancer is rapidly moving forward in multiple areas, including the adoptive transfer of anti-tumor-reactive T cells and the use of "therapeutic" vaccines. The over-expression of RhoC in cancer and the fact that immune escape by down regulation or loss of expression of this protein would reduce the morbidity and mortality of cancer makes RhoC a very attractive target for anti-cancer immunotherapy. Herein, we describe an HLA-A3 restricted epitope from RhoC, which is recognized by cytotoxic T cells. Moreover, RhoC-specific T cells show cytotoxic potential against HLA-matched cancer cells of different origin. Thus, RhoC may serve as an important and widely applicable target for anti-cancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Lynn Wenandy
- Center for Cancer Immune therapy (CCIT), Department of Hematology, Herlev University Hospital, 2730, Herlev, Denmark
| | - Rikke Bæk Sørensen
- Center for Cancer Immune therapy (CCIT), Department of Hematology, Herlev University Hospital, 2730, Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune therapy (CCIT), Department of Hematology, Herlev University Hospital, 2730, Herlev, Denmark
| | - Per thor Straten
- Center for Cancer Immune therapy (CCIT), Department of Hematology, Herlev University Hospital, 2730, Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune therapy (CCIT), Department of Hematology, Herlev University Hospital, 2730, Herlev, Denmark
| |
Collapse
|
17
|
Immunity to Growth Factor Receptor–Bound Protein 10, a Signal Transduction Molecule, Inhibits the Growth of Breast Cancer in Mice. Cancer Res 2008; 68:2463-70. [DOI: 10.1158/0008-5472.can-07-5685] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Brodzik R, Spitsin S, Golovkin M, Bandurska K, Portocarrero C, Okulicz M, Steplewski Z, Koprowski H. Plant-derived EpCAM antigen induces protective anti-cancer response. Cancer Immunol Immunother 2008; 57:317-23. [PMID: 17634938 PMCID: PMC11030716 DOI: 10.1007/s00262-007-0366-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 06/28/2007] [Indexed: 11/28/2022]
Abstract
Immunotherapy holds great promise for treatment of infectious and malignant diseases and might help to prevent the occurrence and recurrence of cancer. We produced a plant-derived tumor-associated colorectal cancer antigen EpCAM (pGA733) at high yields using two modern plant expression systems. The full antigenic domain of EpCAM was efficiently purified to confirm its antigenic and immunogenic properties as compared to those of the antigen expressed in the baculovirus system (bGA733). Recombinant plant-derived antigen induced a humoral immune response in BALB/c mice. Sera from those mice efficiently inhibited the growth of SW948 colorectal carcinoma cells xenografted in nude mice, as compared to the EpCAM-specific mAb CO17-1A. Our results support the feasibility of producing anti-cancer recombinant vaccines using plant expression systems.
Collapse
MESH Headings
- Animals
- Antibodies/blood
- Antibodies, Monoclonal/pharmacology
- Antibody Affinity/immunology
- Antibody Specificity/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/isolation & purification
- Beta vulgaris/genetics
- Beta vulgaris/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/isolation & purification
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/therapy
- Epithelial Cell Adhesion Molecule
- Gene Expression Regulation, Plant/genetics
- Humans
- Immune Sera/pharmacology
- Immunotherapy/methods
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Plants, Genetically Modified/chemistry
- Plants, Genetically Modified/genetics
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Recombinant Proteins/isolation & purification
- Nicotiana/genetics
- Nicotiana/immunology
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Robert Brodzik
- Biotechnology Foundation Laboratories, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107 USA
| | - Sergei Spitsin
- Biotechnology Foundation Laboratories, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107 USA
| | - Max Golovkin
- Biotechnology Foundation Laboratories, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107 USA
| | - Katarzyna Bandurska
- Biotechnology Foundation Laboratories, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107 USA
| | - Carla Portocarrero
- Biotechnology Foundation Laboratories, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107 USA
| | - Monika Okulicz
- Present Address: Department of Animal Physiology and Biochemistry, August Cieszkowski Agricultural University, Wolynska 35, 60-637 Poznan, Poland
| | - Zenon Steplewski
- Biotechnology Foundation Laboratories, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107 USA
| | - Hilary Koprowski
- Biotechnology Foundation Laboratories, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA 19107 USA
| |
Collapse
|
19
|
Skak K, Kragh M, Hausman D, Smyth MJ, Sivakumar PV. Interleukin 21: combination strategies for cancer therapy. Nat Rev Drug Discov 2008; 7:231-40. [DOI: 10.1038/nrd2482] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Karan D, Thrasher JB, Lubaroff D. Prostate cancer: genes, environment, immunity and the use of immunotherapy. Prostate Cancer Prostatic Dis 2008; 11:230-6. [PMID: 18283297 DOI: 10.1038/pcan.2008.3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prostate cancer remains the most prevalent noncutaneous cancer, leading to almost 30,000 deaths every year in men in the United States. A large body of knowledge emphasizes a strong influence of epidemiological factors such as lifestyle, environment and diet, on the development of prostate cancer. Although risk reduction of prostate cancer has been somewhat successful, effective prevention is still lacking. Immunotherapeutic approaches, although moderately complicated, remain promising in an effort to control the progression and development of the disease. Taken together, the parameters of epidemiological studies and immunotherapeutic regimens might eventually be the most effective and preventive approach for prostate cancer. This review highlights some of the events associated with the development and prevention of prostate cancer.
Collapse
Affiliation(s)
- D Karan
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | | | |
Collapse
|
21
|
Kim J, Raz D, Jablons D. Unmet Need in Lung Cancer: Can Vaccines Bridge the Gap? Clin Lung Cancer 2008; 9 Suppl 1:S6-12. [DOI: 10.3816/clc.2008.s.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
22
|
Håkansson L, Dalgleish A, Kallen K, Schuessler-Lenz M. Vaccines: A rapidly evolving technology – Are the hurdles being addressed? EJC Suppl 2007. [DOI: 10.1016/j.ejcsup.2007.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
23
|
Karan D, Krieg AM, Lubaroff DM. Paradoxical enhancement of CD8 T cell-dependent anti-tumor protection despite reduced CD8 T cell responses with addition of a TLR9 agonist to a tumor vaccine. Int J Cancer 2007; 121:1520-8. [PMID: 17565748 DOI: 10.1002/ijc.22873] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Generation of antigen-specific CD8+ T cell responses is considered optimal for an effective immunotherapy against cancer. In this study, we provide a proof of principle that in vitro observed diminished CD8+ T cell response provided a strong in vivo tumor protection. Immunization with an adenovirus vaccine containing ovalbumin (OVA) gene (Ad5-OVA) strongly induces antigen-specific CD8+ T cell responses measured in vitro using various immunological assays. However, in an attempt to augment the antigenic CD8+ T cell response, coinjection of a TLR9 agonist CpG ODN with the viral vaccine unexpectedly reduced the CD8+ T cell responses measured in vitro but provided a remarkably enhanced tumor protection compared to the CD8+ T cell response generated by Ad5-OVA vaccine alone. Interestingly, despite reduced ex vivo/in vitro CD8+ T cell responses following Ad5-OVA+CpG immunization, immunodepletion studies revealed that the augmented anti-tumor immunity was primarily dependent on CD8+ T cells. The magnitude and effector function of anti-OVA CD8+ T cells remain low following primary and secondary antigenic challenge, presenting a dichotomy between in vitro CD8 T cell responses and in vivo anti-tumor immunity. To examine the impact of CpG ODN, we observed that presence of CpG suppresses the CD8+ T cell proliferation both in vitro and in vivo. These data demonstrate that coadministration of adenovirus vaccine with a TLR9 agonist can generate potentially effective tumor-reactive CD8+ T cells in vivo. In addition, the results indicate that widely used standard immune parameters may not predict the vaccine efficacy containing a TLR9 agonist as adjuvant.
Collapse
Affiliation(s)
- Dev Karan
- VA Medical Center, Iowa City, IA, USA
| | | | | |
Collapse
|
24
|
Vuk-Pavlovic S. Rebuilding immunity in cancer patients. Blood Cells Mol Dis 2007; 40:94-100. [PMID: 17827037 PMCID: PMC2225479 DOI: 10.1016/j.bcmd.2007.06.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 06/30/2007] [Accepted: 06/30/2007] [Indexed: 10/22/2022]
Abstract
Rebuilding and maintaining immunity are paramount to the success of cancer immunotherapy and hematopoietic stem cell transplantation. If immune surveillance indeed can protect from cancer, the very manifestation of malignancy means that the disease has prevailed over immunity. Yet, often, tumor-specific T cells can be found in cancer patients irrespective of vaccination. Interestingly, patients suffering from malignancy often harbor unexpectedly high levels of immature CD14(+)HLA-DR(-) monocytes, although the abundance of CD4(+) cells, CD8(+) cells and CD4(+)CD25(high) cells may be normal. It is plausible that in cancer such cells suppress T cell function, analogous to CD14(+)HLA-DR(-) cells in sepsis and major trauma, in addition to their likely failure to re-present tumor-associated antigens once dendritic cells have initiated the T cell response. Recent evidence indicates that tumor-borne adenosine, lactate and hypoxia in the tumor environment may modulate tumor-specific immunity to a significant extent, but their effects on myeloid cell function are unclear. Thus, understanding and controlling these factors may appreciably impact the success of rebuilding and maintaining immunity in cancer patients.
Collapse
Affiliation(s)
- Stanimir Vuk-Pavlovic
- Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
| |
Collapse
|
25
|
O-Sullivan I, Chopra A, Kim TS, Magnuson S, Falduto MT, Huang J, Cohen EP. New strategy for the identification of squamous carcinoma antigens that induce therapeutic immune responses in tumor-bearing mice. Cancer Gene Ther 2007; 14:389-98. [PMID: 17273183 DOI: 10.1038/sj.cgt.7701023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This study describes a new strategy for the identification of squamous carcinoma antigens tumor-associated antigens (TAA). The antigens were discovered by comparing microarrays of squamous carcinoma vaccines highly enriched for immunotherapeutic cells with non-enriched vaccines. The vaccines were prepared by transferring sheared genomic DNA fragments (25 kb) from KLN205 cells, a squamous carcinoma cell line (DBA/2 mouse origin (H-2(d)) into LM fibroblasts (C3H/He origin, H-2(k)). The transferred tumor DNA segments integrate spontaneously into the genome of the recipient cells, replicate as the cells divide and are expressed. As only a small proportion of the transfected cell population was expected to have incorporated DNA segments that included genes specifying TAA (the vast majority specify normal cellular constituents), a novel strategy was employed to enrich the vaccine for TAA-positive cells. Microarrays were used to compare genes expressed by enriched and non-enriched vaccines. Seventy-five genes were overexpressed in cells from the enriched vaccine. One, the gene for Cytochrome P450 (family 2, subfamily e, polypeptide 1) (Cyp2e1), was overexpressed in the enriched but not the non-enriched vaccine. A vaccine for squamous carcinoma was prepared by transfer of a 357 bp fragment of the gene for Cyp2e1 into the fibroblast cell line. Robust immunity, sufficient to result in indefinite survival, was induced in tumor-bearing mice immunized with cells transfected with this gene fragment.
Collapse
Affiliation(s)
- I O-Sullivan
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60615, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Listner K, Bentley L, Okonkowski J, Kistler C, Wnek R, Caparoni A, Junker B, Robinson D, Salmon P, Chartrain M. Development of a highly productive and scalable plasmid DNA production platform. Biotechnol Prog 2007; 22:1335-45. [PMID: 17022672 DOI: 10.1021/bp060046h] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
With the applications of DNA vaccines extending from infectious diseases to cancer, achieving the most efficient, reproducible, robust, scalable, and economical production of clinical grade plasmid DNA is paramount to the medical and commercial success of this novel vaccination paradigm. A first generation production process based on the cultivation of Escherichia coli in a chemically defined medium, employing a fed-batch strategy, delivered reasonable volumetric productivities (500-750 mg/L) and proved to perform very well across a wide range of E. coli constructs upon scale-up at industrial scale. However, the presence of monosodium glutamate (MSG) in the formulation of the cultivation and feed solution was found to be a potential cause of process variability. The development of a second generation process, based on a defined cultivation medium and feed solution excluding MSG, was undertaken. Optimization studies, employing a plasmid coding for the HIV gag protein, resulted in cultivation conditions that supported volumetric plasmid titers in excess of 1.2 g/L, while achieving specific yields ranging from 25 to 32 microg plasmid DNA/mg of dry cell weight. When used for the production of clinical supplies, this novel process demonstrated applicability to two other constructs upon scale-up in 2,000-L bioreactors. This second generation process proved to be scalable, robust, and highly productive.
Collapse
Affiliation(s)
- K Listner
- Bioprocess R&D, Merck Research Laboratories, Rahway, New Jersey 07065, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Durrant LG. Conference report and introduction to the symposium papers. Cancer Immunol Immunother 2006. [DOI: 10.1007/s00262-006-0144-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|