1
|
Smith R, Wafa EI, Geary SM, Ebeid K, Alhaj-Suliman SO, Salem AK. Cationic nanoparticles enhance T cell tumor infiltration and antitumor immune responses to a melanoma vaccine. SCIENCE ADVANCES 2022; 8:eabk3150. [PMID: 35857851 PMCID: PMC9299550 DOI: 10.1126/sciadv.abk3150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
In clinical settings, cancer vaccines as monotherapies have displayed limited success compared to other cancer immunotherapeutic treatments. Nanoscale formulations have the ability to increase the efficacy of cancer vaccines by combatting the immunosuppressive nature of the tumor microenvironment. Here, we have synthesized a previously unexplored cationic polymeric nanoparticle formulation using polyamidoamine dendrimers and poly(d,l-lactic-co-glycolic acid) that demonstrate adjuvant properties in vivo. Tumor-challenged mice vaccinated with an adenovirus-based cancer vaccine [encoding tumor-associated antigen (TAA)] and subsequently treated with this nanoparticulate formulation showed significant increases in TAA-specific T cells in the peripheral blood, reduced tumor burden, protection against tumor rechallenge, and a significant increase in median survival. An investigation into cell-based pathways suggests that administration of the nanoformulation at the site of the developing tumor may have created an inflammatory environment that attracted activated TAA-specific CD8+ T cells to the vicinity of the tumor, thus enhancing the efficacy of the vaccine.
Collapse
Affiliation(s)
| | | | | | - Kareem Ebeid
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Suhaila O. Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
2
|
Movassaghi M, Chung R, Anderson CB, Stein M, Saenger Y, Faiena I. Overcoming Immune Resistance in Prostate Cancer: Challenges and Advances. Cancers (Basel) 2021; 13:cancers13194757. [PMID: 34638243 PMCID: PMC8507531 DOI: 10.3390/cancers13194757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Immunotherapy has changed the landscape of treatment modalities available for many different types of malignancies. However, the factors that influence the success of immunotherapeutics have not been as clearly seen in advanced prostate cancer, likely due to immunosuppressive factors that exist within the prostate cancer tumor microenvironment. While there have been many immunotherapeutics used for prostate cancer, the majority have targeted a single immunosuppressive mechanism resulting in limited clinical efficacy. More recent research centered on elucidating the key mechanisms of immune resistance in the prostate tumor microenvironment has led to the discovery of a range of new treatment targets. With that in mind, many clinical trials have now set out to evaluate combination immunotherapeutic strategies in patients with advanced prostate cancer, in the hopes of circumventing the immunosuppressive mechanisms. Abstract The use of immunotherapy has become a critical treatment modality in many advanced cancers. However, immunotherapy in prostate cancer has not been met with similar success. Multiple interrelated mechanisms, such as low tumor mutational burden, immunosuppressive cells, and impaired cellular immunity, appear to subvert the immune system, creating an immunosuppressive tumor microenvironment and leading to lower treatment efficacy in advanced prostate cancer. The lethality of metastatic castrate-resistant prostate cancer is driven by the lack of therapeutic regimens capable of generating durable responses. Multiple strategies are currently being tested to overcome immune resistance including combining various classes of treatment modalities. Several completed and ongoing trials have shown that combining vaccines or checkpoint inhibitors with hormonal therapy, radiotherapy, antibody–drug conjugates, chimeric antigen receptor T cell therapy, or chemotherapy may enhance immune responses and induce long-lasting clinical responses without significant toxicity. Here, we review the current state of immunotherapy for prostate cancer, as well as tumor-specific mechanisms underlying therapeutic resistance, with a comprehensive look at the current preclinical and clinical immunotherapeutic strategies aimed at overcoming the immunosuppressive tumor microenvironment and impaired cellular immunity that have largely limited the utility of immunotherapy in advanced prostate cancer.
Collapse
Affiliation(s)
- Miyad Movassaghi
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
- Correspondence: (M.M.); (I.F.)
| | - Rainjade Chung
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
| | - Christopher B. Anderson
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
| | - Mark Stein
- Department of Medicine, Division of Medical Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (Y.S.)
| | - Yvonne Saenger
- Department of Medicine, Division of Medical Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (Y.S.)
| | - Izak Faiena
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
- Correspondence: (M.M.); (I.F.)
| |
Collapse
|
3
|
Adamaki M, Zoumpourlis V. Immunotherapy as a Precision Medicine Tool for the Treatment of Prostate Cancer. Cancers (Basel) 2021; 13:E173. [PMID: 33419051 PMCID: PMC7825410 DOI: 10.3390/cancers13020173] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed type of cancer among Caucasian males over the age of 60 and is characterized by remarkable heterogeneity and clinical behavior, ranging from decades of indolence to highly lethal disease. Despite the significant progress in PCa systemic therapy, therapeutic response is usually transient, and invasive disease is associated with high mortality rates. Immunotherapy has emerged as an efficacious and non-toxic treatment alternative that perfectly fits the rationale of precision medicine, as it aims to treat patients on the basis of patient-specific, immune-targeted molecular traits, so as to achieve the maximum clinical benefit. Antibodies acting as immune checkpoint inhibitors and vaccines entailing tumor-specific antigens seem to be the most promising immunotherapeutic strategies in offering a significant survival advantage. Even though patients with localized disease and favorable prognostic characteristics seem to be the ones that markedly benefit from such interventions, there is substantial evidence to suggest that the survival benefit may also be extended to patients with more advanced disease. The identification of biomarkers that can be immunologically targeted in patients with disease progression is potentially amenable in this process and in achieving significant advances in the decision for precision treatment of PCa.
Collapse
Affiliation(s)
- Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece;
| | | |
Collapse
|
4
|
Handa S, Hans B, Goel S, Bashorun HO, Dovey Z, Tewari A. Immunotherapy in prostate cancer: current state and future perspectives. Ther Adv Urol 2020; 12:1756287220951404. [PMID: 32952615 PMCID: PMC7476347 DOI: 10.1177/1756287220951404] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/25/2020] [Indexed: 02/06/2023] Open
Abstract
Metastatic castrate resistant prostate cancer (PCa) remains an incurable entity. In the era of immunotherapy, the complex PCa microenvironment poses a unique challenge to the successful application of this class of agents. However, in the last decade, a tremendous effort has been made to explore this field of therapeutics. In this review, the physiology of the cancer immunity cycle is highlighted in the context of the prostate tumor microenvironment, and the current evidence for use of various classes of immunotherapy agents including vaccines (dendritic cell based, viral vector based and DNA/mRNA based), immune checkpoint inhibitors, Chimeric antigen receptor T cell therapy, antibody-mediated radioimmunotherapy, antibody drug conjugates, and bispecific antibodies, is consolidated. Finally, the future directions for combinatorial approaches to combat PCa are discussed.
Collapse
Affiliation(s)
- Shivani Handa
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West Hospital, New York, NY, 10019, USA
| | - Bandhul Hans
- Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Shokhi Goel
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Hafis O Bashorun
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Zach Dovey
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Ashutosh Tewari
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
5
|
Hossain MK, Nahar K, Donkor O, Apostolopoulos V. Immune-based therapies for metastatic prostate cancer: an update. Immunotherapy 2019; 10:283-298. [PMID: 29421982 DOI: 10.2217/imt-2017-0123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer (PC) is a common malignancy among elderly males and is noncurable once it becomes metastatic. In recent years, a number of antigen-delivery systems have emerged as viable and promising immunotherapeutic agents against PC. The approval of sipuleucel-T by the US FDA for the treatment of males with asymptomatic or minimally symptomatic castrate resistant PC was a landmark in cancer immunotherapy, making this the first approved immunotherapeutic. A number of vaccines are under clinical investigation, each having its own set of advantages and disadvantages. Here, we discuss the basic technologies underlying these different delivery modes, we discuss the completed and current human clinical trials, as well as the use of vaccines in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Kamrun Nahar
- Vetafarm Pty Ltd, Wagga Wagga, NSW, 2650, Australia
| | - Osaana Donkor
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Australia
| | - Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Australia
| |
Collapse
|
6
|
Abstract
Polymeric matrices inherently protect viral vectors from pre-existing immune conditions, limit dissemination to off-target sites, and can sustain vector release. Advancing methodologies in development of particulate based vehicles have led to improved encapsulation of viral vectors. Polymeric delivery systems have contributed to increasing cellular transduction, responsive release mechanisms, cellular infiltration, and cellular signaling. Synthetic polymers are easily customizable, and are capable of balancing matrix retention with cellular infiltration. Natural polymers contain inherent biorecognizable motifs adding therapeutic efficacy to the incorporated viral vector. Recombinant polymers use highly conserved motifs to carefully engineer matrices, allowing for precise design including elements of vector retention and responsive release mechanisms. Composite polymer systems provide opportunities to create matrices with unique properties. Carefully designed matrices can control spatiotemporal release patterns that synergize with approaches in regenerative medicine and antitumor therapies.
Collapse
Affiliation(s)
- Douglas Steinhauff
- Utah Center for Nanomedicine , Nano Institute of Utah , 36 South Wasatch Drive , Salt Lake City , Utah 84112 , United States
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine , Nano Institute of Utah , 36 South Wasatch Drive , Salt Lake City , Utah 84112 , United States
| |
Collapse
|
7
|
Formulation of the bivalent prostate cancer vaccine with surgifoam elicits antigen-specific effector T cells in PSA-transgenic mice. Vaccine 2017; 35:5794-5798. [PMID: 28939158 DOI: 10.1016/j.vaccine.2017.09.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/07/2017] [Accepted: 09/12/2017] [Indexed: 11/24/2022]
Abstract
We previously developed and characterized an adenoviral-based prostate cancer vaccine for simultaneous targeting of prostate-specific antigen (PSA) and prostate stem cell antigen (PSCA). We also demonstrated that immunization of mice with the bivalent vaccine (Ad5-PSA+PSCA) inhibited the growth of established prostate tumors. However, there are multiple challenges hindering the success of immunological therapies in the clinic. One of the prime concerns has been to overcome the immunological tolerance and maintenance of long-term effector T cells. In this study, we further characterized the use of the bivalent vaccine (Ad5-PSA+PSCA) in a transgenic mouse model expressing human PSA in the mouse prostate. We demonstrated the expression of PSA analyzed at the mRNA level (by RT-PCR) and protein level (by immunohistochemistry) in the prostate lobes harvested from the PSA-transgenic (PSA-Tg) mice. We established that the administration of the bivalent vaccine in surgifoam to the PSA-Tg mice induces strong PSA-specific effector CD8+ T cells as measured by IFN-γ secretion and in vitro cytotoxic T-cell assay. Furthermore, the use of surgifoam with Ad5-PSA+PSCA vaccine allows multiple boosting vaccinations with a significant increase in antigen-specific CD8+ T cells. These observations suggest that the formulation of the bivalent prostate cancer vaccine (Ad5-PSA+PSCA) with surgifoam bypasses the neutralizing antibody response, thus allowing multiple boosting. This formulation is also helpful for inducing an antigen-specific immune response in the presence of self-antigen, and maintains long-term effector CD8+ T cells.
Collapse
|
8
|
Hadac JN, Leystra AA, Paul Olson TJ, Maher ME, Payne SN, Yueh AE, Schwartz AR, Albrecht DM, Clipson L, Pasch CA, Matkowskyj KA, Halberg RB, Deming DA. Colon Tumors with the Simultaneous Induction of Driver Mutations in APC, KRAS, and PIK3CA Still Progress through the Adenoma-to-carcinoma Sequence. Cancer Prev Res (Phila) 2015; 8:952-61. [PMID: 26276752 DOI: 10.1158/1940-6207.capr-15-0003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 07/15/2015] [Indexed: 02/06/2023]
Abstract
Human colorectal cancers often possess multiple mutations, including three to six driver mutations per tumor. The timing of when these mutations occur during tumor development and progression continues to be debated. More advanced lesions carry a greater number of driver mutations, indicating that colon tumors might progress from adenomas to carcinomas through the stepwise accumulation of mutations following tumor initiation. However, mutations that have been implicated in tumor progression have been identified in normal-appearing epithelial cells of the colon, leaving the possibility that these mutations might be present before the initiation of tumorigenesis. We utilized mouse models of colon cancer to investigate whether tumorigenesis still occurs through the adenoma-to-carcinoma sequence when multiple mutations are present at the time of tumor initiation. To create a model in which tumors could concomitantly possess mutations in Apc, Kras, and Pik3ca, we developed a novel minimally invasive technique to administer an adenovirus expressing Cre recombinase to a focal region of the colon. Here, we demonstrate that the presence of these additional driver mutations at the time of tumor initiation results in increased tumor multiplicity and an increased rate of progression to invasive adenocarcinomas. These cancers can even metastasize to retroperitoneal lymph nodes or the liver. However, despite having as many as three concomitant driver mutations at the time of initiation, these tumors still proceed through the adenoma-to-carcinoma sequence.
Collapse
Affiliation(s)
- Jamie N Hadac
- Department of Oncology, University of Wisconsin, Madison, Wisconsin
| | - Alyssa A Leystra
- Department of Oncology, University of Wisconsin, Madison, Wisconsin
| | - Terrah J Paul Olson
- Division of General Surgery, Department of Surgery, University of Wisconsin, Madison, Wisconsin
| | - Molly E Maher
- Division of Hematology and Oncology, University of Wisconsin, Madison, Wisconsin
| | - Susan N Payne
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Alexander E Yueh
- Division of Hematology and Oncology, University of Wisconsin, Madison, Wisconsin
| | - Alexander R Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Wisconsin, Madison, Wisconsin
| | - Dawn M Albrecht
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Wisconsin, Madison, Wisconsin
| | - Linda Clipson
- Department of Oncology, University of Wisconsin, Madison, Wisconsin
| | - Cheri A Pasch
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Kristina A Matkowskyj
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin. William S Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Richard B Halberg
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. Division of Gastroenterology and Hepatology, Department of Medicine, University of Wisconsin, Madison, Wisconsin
| | - Dustin A Deming
- Division of Hematology and Oncology, University of Wisconsin, Madison, Wisconsin. University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin. William S Middleton Memorial Veterans Hospital, Madison, Wisconsin.
| |
Collapse
|
9
|
Lubaroff DM, Vaena D, Brown JA, Zehr P, Griffith KC, Brown E, Eastman J, Nepple K, Kattula A, Williams RD. Vaccine immunotherapy for prostate cancer: from mice to men. Immunol Res 2015; 59:229-35. [PMID: 24847764 DOI: 10.1007/s12026-014-8531-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Preclinical studies demonstrated the ability of an adenovirus/PSA (Ad/PSA) vaccine to induce strong anti-PSA immune responses, and these responses were capable of destroying prostate-specific antigen (PSA)-secreting mouse prostate tumors. A series of preclinical studies have demonstrated the superiority of the Ad/PSA vaccine to other PSA vaccines for the induction of anti-PSA immune responses, the ability of Ad/PSA vaccination combined with cytokine gene therapy and the TLR9 agonist CpG to enhance the anti-prostate tumor immunotherapy, and the reduction of negative regulatory elements when the vaccine was combined with 5-fluoruracil administration. A phase I clinical trial of the Ad/PSA vaccine in men with metastatic castrate-resistant prostate cancer demonstrated the safety of the vaccine even at the highest single dose permitted by the FDA. Currently, a phase II trial of the Ad/PSA vaccine is underway treating patients in two protocols. Thus far 81 patients have been enrolled and vaccinated. Early results from the patients evaluated to date demonstrated the induction of anti-PSA T cell responses, and the majority of patients evaluated at this time had demonstrated an increase in PSA doubling times.
Collapse
Affiliation(s)
- David M Lubaroff
- Department of Urology, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Fernández-García EM, Vera-Badillo FE, Perez-Valderrama B, Matos-Pita AS, Duran I. Immunotherapy in prostate cancer: review of the current evidence. Clin Transl Oncol 2014; 17:339-57. [DOI: 10.1007/s12094-014-1259-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 11/21/2014] [Indexed: 01/03/2023]
|
11
|
|
12
|
The combination of a low-dose chemotherapeutic agent, 5-fluorouracil, and an adenoviral tumor vaccine has a synergistic benefit on survival in a tumor model system. PLoS One 2013; 8:e67904. [PMID: 23840786 PMCID: PMC3695864 DOI: 10.1371/journal.pone.0067904] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/23/2013] [Indexed: 12/13/2022] Open
Abstract
Standard cancer therapies, particularly those involving chemotherapy, are in need of modifications that both reduce short-term and long-term side effects as well as improve the overall survival of cancer patients. Here we show that combining low-dose chemotherapy with a therapeutic vaccination using an adenovirus encoding a model tumor-associated antigen, ovalbumin (Ad5-OVA), had a synergistic impact on survival in tumor-challenged mice. Mice that received the combinatorial treatment of Ad5-OVA plus low-dose 5-fluorouracil (5-FU) had a 95% survival rate compared to 7% and 30% survival rates for Ad5-OVA alone and 5-FU alone respectively. The presence of 5-FU enhanced the levels of OVA-specific CD8+ T lymphocytes in the spleens and draining lymph nodes of Ad5-OVA-treated mice, a phenomenon that was dependent on the mice having been tumor-challenged. Thus 5-FU may have enhanced survival of Ad5-OVA-treated mice by enhancing the tumor-specific immune response combined with eliminating tumor bulk. We also investigated the possibility that the observed therapeutic benefit may have been derived from the capacity of 5-FU to deplete MDSC populations. The findings presented here promote the concept of combining adenoviral cancer vaccines with low-dose chemotherapy.
Collapse
|
13
|
Geary SM, Salem AK. Prostate cancer vaccines: Update on clinical development. Oncoimmunology 2013; 2:e24523. [PMID: 23762812 PMCID: PMC3667918 DOI: 10.4161/onci.24523] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 04/02/2013] [Indexed: 01/05/2023] Open
Abstract
Prostate cancer is a common malignancy among elderly men and is essentially incurable once it becomes metastatic. Results from clinical trials testing a panel of specific vaccines in patients with castration-resistant prostate cancer (CRPC) suggest that alternative therapies may one day substitute or support the current gold standard (docetaxel plus prednisone). Here, we summarize the results of germane clinical trials completed during the last 12 y and provide updates on some currently ongoing studies. As it stands, prostate cancer vaccines appear to be safe and capable of generating prostate-specific T lymphocyte responses with potential antitumor activity.
Collapse
Affiliation(s)
- Sean M Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics; College of Pharmacy; University of Iowa; Iowa City, IA USA
| | | |
Collapse
|
14
|
Abstract
Prostate cancer is responsible for the deaths of more than 33,000 American men every year. Once this disease has become metastatic, there is no curative treatment. Alternative therapies to chemotherapy and radical prostatectomy are being increasingly explored. Prostate cancer vaccines--which trigger a tumour-specific cytotoxic-T-lymphocyte-mediated immune attack by the patient's immune system--have been investigated in clinical trials with modest, yet encouraging, results. When developing and administering prostate cancer vaccines, it is critical to consider how vital parameters, such as the stage of disease progression and the nature of adjuvant therapies, could influence treatment outcome. Of particular interest are current and future strategies for diminishing the activity of regulatory T lymphocytes.
Collapse
|
15
|
Antigen-coated poly α-hydroxy acid based microparticles for heterologous prime-boost adenovirus based vaccinations. Biomaterials 2013; 34:2524-9. [PMID: 23312902 DOI: 10.1016/j.biomaterials.2012.12.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/22/2012] [Indexed: 02/06/2023]
Abstract
Adenoviruses show promising potential as vectors for cancer vaccines, however, their high immunogenicity can be problematic when it comes to homologous prime-boost strategies. In the studies presented here we show that heterologous prime-boost vaccinations involving ovalbumin (OVA)-antigen-coated microparticles as a prime, and adenovirus encoding OVA (AdOVA) as a boost, were equally as effective as homologous AdOVA prime-boosts at generating OVA-specific CD8(+) T-cell responses, which translated into effective tumor protection. OVA-coated biodegradable poly α-hydroxy acid-based microparticles of varying chemistries, when used as primes in heterologous prime-boost vaccinations, were comparable in terms of promoting OVA-specific CD8(+) T cells as well as providing protection against subsequent tumor challenge. These findings auger well for using poly α-hydroxy acid-based microparticles in prime-boost viral vaccination strategies geared toward the safer, and potentially more efficient, generation of anti-tumor immunity.
Collapse
|
16
|
Abstract
The improved survival with sipuleucel-T, an autologous antigen-presenting cell-based agent, for the treatment of patients with metastatic asymptomatic and minimally symptomatic castration-resistant prostate cancer supports immunotherapy as a valid approach. Also, multiple novel immunotherapeutic approaches are undergoing vigorous investigation. T-lymphocyte checkpoint blockade and poxvirus-based prime-boost approaches are in phase III evaluation. Other immunotherapeutic platforms undergoing early investigation include radioimmunoconjugates and adenovirus-based, DNA-based, and Listeria-based approaches. The development of predictive markers for immune response that translate into improved long-term outcomes is important. This article reviews the emerging data and the unique strengths and weaknesses of these approaches.
Collapse
Affiliation(s)
- Guru Sonpavde
- Department of Medicine, Section of Medical Oncology, University of Alabama at Birmingham (UAB) Comprehensive Cancer Center, Birmingham, AL 35294, USA
| | | |
Collapse
|
17
|
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
|
18
|
Abstract
The recent approval of Sipuleucel-T (Dendreon, Seattle, WA) from the Food and Drug Administration for the treatment of men with asymptomatic or minimally symptomatic castrate-resistant prostate cancer was a landmark in cancer immunotherapy, making this the first cancer "vaccine" approved for use in a treatment setting. This approval has led to renewed interest in cancer vaccines and to the recognition that prostate cancer represents an immunologically sensitive disease. At the current time, several vaccine approaches are under clinical investigation. These include viral vectors, antigen-loaded dendritic cells, and DNA vaccines. Each approach has its own set of advantages and disadvantages. This review will introduce the basic technology underlying these different vaccines and briefly discuss completed and ongoing clinical trials. As a great number of prostate cancer vaccines have been investigated in both preclinical and clinical settings, we will focus primarily on vaccines that are currently in clinical trials, as ascertained by a recent inquiry of the clinical trials database, www.clinicaltrials.gov.
Collapse
|
19
|
Lemke CD, Graham JB, Geary SM, Zamba G, Lubaroff DM, Salem AK. Chitosan is a surprising negative modulator of cytotoxic CD8+ T cell responses elicited by adenovirus cancer vaccines. Mol Pharm 2011; 8:1652-61. [PMID: 21780831 DOI: 10.1021/mp100464y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adjuvants modulate protective CD8(+) T cell responses generated by cancer vaccines. We have previously shown that immunostimulatory cytosine-phosphodiester-guanine (CpG) oligodeoxynucleotide (ODN) significantly augments tumor protection in mice given adenovirus cancer vaccines. Here, we examined the impact of chitosan, another candidate vaccine adjuvant, on protection conferred by adenovirus cancer vaccines. Unexpectedly, immunization of mice with adenovirus cancer vaccines in combination with chitosan provided little protection against tumor challenge. This directly correlated with the reduced detection of Ag-specific CD8(+) T cells, interferon-γ (IFN-γ) production, and cytotoxic T cell activity. We ruled out immunosuppressive regulatory T cells since the frequency did not change regardless of whether chitosan was delivered. In mammalian cell lines, chitosan did not interfere with adenovirus transgene expression. However, infection of primary murine bone marrow-derived dendritic cells with adenovirus complexed with chitosan significantly reduced viability, transgene expression, and upregulation of major histocompatability (MHC) class I and CD86. Our in vitro observations indicate that chitosan dramatically inhibits adenovirus-mediated transgene expression and antigen presenting cell activation, which could prevent CD8(+) T cell activation from occurring in vivo. These surprising data demonstrate for the first time that chitosan vaccine formulations can negatively impact the induction of CD8(+) T cell responses via its effect on dendritic cells, which is clinically important since consideration of chitosan as an adjuvant for vaccine formulations is growing.
Collapse
Affiliation(s)
- Caitlin D Lemke
- College of Pharmacy, College of Public Health, University of Iowa, Iowa City, Iowa 52242, United States
| | | | | | | | | | | |
Collapse
|
20
|
Shanmugam A, Suriano R, Chaudhuri D, Rajoria S, George A, Mittelman A, Tiwari RK. Identification of PSA peptide mimotopes using phage display peptide library. Peptides 2011; 32:1097-102. [PMID: 21539876 DOI: 10.1016/j.peptides.2011.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 04/15/2011] [Accepted: 04/16/2011] [Indexed: 11/21/2022]
Abstract
Prostate cancer (PCa) is one of the most common types of cancer in men in the United States and is the second leading cause of cancer related death in men. Clinically, secreted prostate specific antigen (PSA) has gained recognition because of its proteolytic activity being directly linked to PCa cell proliferation leading to disease initiation and progression. Using phage display technology, we identified four distinct cyclical peptides. These peptides apart from differences in their amino acid sequence, elicited minimal cross reactive antibody responses against each other. One of the four peptides analyzed produced an antibody response that recognizes the PSA protein. We demonstrate that the synthetic PSA peptide mimics identified in our study are immunologically active and produce neutralizing activity and this has relevance and utility for prostate cancer disease progression.
Collapse
Affiliation(s)
- Arulkumaran Shanmugam
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Development of an MHC class I L(d)-restricted PSA peptide-loaded tetramer for detection of PSA-specific CD8+ T cells in the mouse. Prostate Cancer Prostatic Dis 2011; 14:118-21. [PMID: 21263453 PMCID: PMC3094480 DOI: 10.1038/pcan.2010.57] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Objectives We set out to develop a prostate specific antigen (PSA) peptide-loaded tetramer for enumeration of PSA-specific CD8+ T cells in the Balb/c mouse model. Methods A candidate MHC class I PSA peptide (HPQKVTKFML188–197) was selected based on its ability to restimulate PSA-specific CD8+ T cells to secrete IFN-γ in our assays. Next, H-2Ld-restricted peptide-loaded and fluorescently labeled tetramers were produced in conjunction with the NIH Tetramer Core Facility. This tetramer was then tested for staining specificity and optimized for detection of PSA-specific CD8+ T cells induced by our PSA-encoding adenovirus tumor vaccine. Results The MHC class I PSA peptide demonstrated successful restimulation of CD8+ T cells isolated from mice previously vaccinated with a PSA-encoding adenovirus tumor vaccine, with no restimulation observed in control vaccinated mice. The peptide-loaded H-2Ld tetramer exhibited the desired binding specificity and allowed for detection and frequency determination of PSA-specific CD8+ T cells by flow cytometry. Conclusions We have successfully designed and validated a PSA peptide tetramer for use in the Balb/c mouse model that can be used to test PSA-based prostate cancer vaccines. Until now, PSA-specific CD8+ T cells in the mouse have only been detectable via cytotoxic T lymphocyte (CTL) assays or intracellular cytokine staining, which primarily assess Ag-specific functional activity, not their absolute number. This research tool provides laboratories the ability to directly quantitate CD8+ T cells elicited by PSA-specific immunotherapies and cancer vaccines that are tested in mouse models.
Collapse
|
22
|
Saito K, Shirasawa H, Isegawa N, Shiiba M, Uzawa K, Tanzawa H. Oncolytic virotherapy for oral squamous cell carcinoma using replication-competent viruses. Oral Oncol 2009; 45:1021-7. [DOI: 10.1016/j.oraloncology.2009.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 08/29/2009] [Accepted: 09/02/2009] [Indexed: 01/02/2023]
|
23
|
Lubaroff DM, Konety BR, Link B, Gerstbrein J, Madsen T, Shannon M, Howard J, Paisley J, Boeglin D, Ratliff TL, Williams RD. Phase I clinical trial of an adenovirus/prostate-specific antigen vaccine for prostate cancer: safety and immunologic results. Clin Cancer Res 2009; 15:7375-80. [PMID: 19920098 DOI: 10.1158/1078-0432.ccr-09-1910] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE We performed a phase I clinical trial of adenovirus/prostate-specific antigen (PSA) vaccine in men with measurable metastatic hormone-refractory disease. EXPERIMENTAL DESIGN Men with measurable metastatic disease received one vaccine injection. Toxicity, immune responses, changes in PSA doubling times, and patient survival were assessed. Thirty-two patients with hormone-refractory metastatic prostate cancer were treated with a single s.c. vaccine injection at one of three dose levels, either as an aqueous solution or suspended in a Gelfoam matrix. All patients returned for physical and clinical chemistry examinations at regular intervals up to 12 months after injections. RESULTS The vaccine was deemed safe at all doses in both administration forms. There were no serious vaccine-related adverse events; the most prevalent were localized erythema/ecchymoses and cold/flu-like symptoms. Anti-PSA antibodies were produced by 34% of patients and anti-PSA T-cell responses were produced by 68%. PSA doubling time was increased in 48%, whereas 55% survived longer than predicted by the Halabi nomogram. CONCLUSIONS The adenovirus/PSA vaccine was proven safe with no serious vaccine-related adverse events. The majority of vaccinated patients produced anti-PSA T-cell responses and over half survived longer than predicted by nomogram. Although the latter data are only derived from a small number of patients in this phase I trial, they are encouraging enough to pursue further studies.
Collapse
Affiliation(s)
- David M Lubaroff
- Department of Urology, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Transcriptionally regulated, prostate-targeted gene therapy for prostate cancer. Adv Drug Deliv Rev 2009; 61:572-88. [PMID: 19393705 DOI: 10.1016/j.addr.2009.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 03/10/2009] [Indexed: 01/08/2023]
Abstract
Prostate cancer is the most frequently diagnosed cancer and the second leading cause of cancer deaths in American males today. Novel and effective treatment such as gene therapy is greatly desired. The early viral based gene therapy uses tissue-nonspecific promoters, which causes unintended toxicity to other normal tissues. In this chapter, we will review the transcriptionally regulated gene therapy strategy for prostate cancer treatment. We will describe the development of transcriptionally regulated prostate cancer gene therapy in the following areas: (1) Comparison of different routes for best viral delivery to the prostate; (2) Study of transcriptionally regulated, prostate-targeted viral vectors: specificity and activity of the transgene under several different prostate-specific promoters were compared in vitro and in vivo; (3) Selection of therapeutic transgenes and strategies for prostate cancer gene therapy (4) Oncolytic virotherapy for prostate cancer. In addition, the current challenges and future directions in this field are also discussed.
Collapse
|
25
|
CpG oligonucleotide as an adjuvant for the treatment of prostate cancer. Adv Drug Deliv Rev 2009; 61:268-74. [PMID: 19166887 DOI: 10.1016/j.addr.2008.12.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2008] [Indexed: 12/18/2022]
Abstract
The use of an adenovirus transduced to express a prostate cancer antigen (PSA) as a vaccine for the treatment of prostate cancer has been shown to be active in the destruction of antigen-expressing prostate tumor cells in a pre-clinical model, using Balb/C or PSA transgenic mice. The destruction of PSA-secreting mouse prostate tumors was observed in Ad/PSA immunized mice in a prophylaxis study with 70% of the mice surviving long term tumor free. This successful immunotherapy was not observed in therapeutic studies in which tumors were established before vaccination and the development of anti-PSA immune response was not as easily generated in PSA transgenic mice. Immunization of conventional and transgenic animals was enhanced by incorporating a collagen matrix into the immunizing injection. Therefore the need to strengthen anti-PSA and anti-prostate cancer immunity was an obvious next step in developing a successful prostate cancer immunotherapy. Because the use of immunostimulatory CpG motifs was shown to enhance immune responses to a wide variety of antigens, our studies incorporated CpG into the Ad/PSA vaccine experimental plans. The results of the subsequent studies demonstrated a dichotomy where Ad/PSA plus CpG enhanced the in vivo destruction of PSA-secreting tumors and the survival of experimental animals, but revealed that the number and in vitro activities of antigen specific CD8+ T cells was decreased as compared to the values observed when the vaccine alone was used for immunization. The dichotomous observations were confirmed using another antigen system, OVA also incorporated into a replication defective adenovirus. Despite the reduction in antigen-specific CD8+ cells after vaccine plus CpG immunization the enhanced destruction of sc and systemic tumors was shown to be mediated entirely by CD8+ T cells. Finally, the reduction of the CD8+ T cells was the result of an observed decrease in the proliferation of the antigen specific cell population.
Collapse
|
26
|
Platelet-derived CD154 enables T-cell priming and protection against Listeria monocytogenes challenge. Blood 2008; 111:3684-91. [PMID: 18256321 DOI: 10.1182/blood-2007-05-091728] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Collagen exposure in tissue activates platelets, initiates wound healing, and modulates adaptive immunity. In this report, data are presented to demonstrate a requirement for platelet-derived CD154 for both collagen-induced augmentation of T-cell immunity and induction of pro-tective immunity to Listeria challenge. Specifically, we demonstrate that Ad5 encoding the membrane-bound form of ovalbumin (Ad5-mOVA) delivered in collagen induces higher ovalbumin-specific cytotoxic T lymphocyte (CTL) activity in a dose-dependent manner compared with Ad5-mOVA delivered in PBS. Increased CTL activity was dependent on the ability of platelets to respond to collagen and to express CD154. Furthermore, mice immunized with low-dose Ad5-mOVA in collagen were able to control a challenge of Listeria monocytogenes recombinant for ovalbumin expression (Lm-OVA), whereas mice immunized with low-dose Ad5-mOVA in PBS were not. These data indicate that in a physiologic setting that mimics wounding, platelets perform a sentinel function when antigen dose is too low to provoke an efficient immune response, and can enhance the generation of antigen-specific CD8 T cells that are functionally relevant to the host.
Collapse
|
27
|
VanOosten RL, Griffith TS. Activation of tumor-specific CD8+ T Cells after intratumoral Ad5-TRAIL/CpG oligodeoxynucleotide combination therapy. Cancer Res 2008; 67:11980-90. [PMID: 18089829 DOI: 10.1158/0008-5472.can-07-1526] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD8(+) T-cell activation via cross-presentation of antigens from apoptotic tumor cells is controversial. Dendritic cells capture naturally shed tumor antigens and cross-present them to CD8(+) T cells; unfortunately, the frequency of activated CD8(+) T cells is often too low to mount an effective response against the tumor. By increasing the amount of antigen for presentation, a larger T-cell response can be theoretically elicited. We used a recombinant adenovirus encoding full-length murine tumor necrosis factor-related apoptosis-inducing ligand (Ad5-mTRAIL) to induce tumor cell apoptosis, and when given intratumorally to mice bearing experimental renal cell carcinoma (Renca) tumors, Ad5-mTRAIL minimally prolonged survival and induced a low level of CTL activity. To enhance dendritic cell efficiency, an immunostimulatory CpG oligodeoxynucleotide (CpG ODN) was combined with Ad5-mTRAIL. This combination therapy significantly augmented in vivo antigen-specific T-cell proliferation and CTL activity, as well as prolonged survival of Renca tumor-bearing mice. Interestingly, depletion of CD4(+) or CD25(+) cells before therapy further enhanced survival and in vivo CTL activity. In addition, tumor-free mice depleted of CD4(+) cells were also able to reject a subsequent challenge of Renca cells, but not MHC-matched RM-11 prostate tumor cells, demonstrating the existence of immunologic memory. These results collectively show that local treatment with Ad5-mTRAIL and CpG ODN can augment tumor antigen cross-presentation resulting in T-cell proliferation, enhanced CTL activity, and increased animal survival.
Collapse
Affiliation(s)
- Rebecca L VanOosten
- Department of Urology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242-1089, USA
| | | |
Collapse
|
28
|
Yang TC, Millar JB, Grinshtein N, Bassett J, Finn J, Bramson JL. T-cell immunity generated by recombinant adenovirus vaccines. Expert Rev Vaccines 2007; 6:347-56. [PMID: 17542750 DOI: 10.1586/14760584.6.3.347] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recombinant adenovirus vaccines show great promise for generating protective immunity against infectious agents and tumors. Our studies have identified several interesting biological features of the adenovirus vector that influence the T-cell response. Notably, we have demonstrated that following immunization with adenovirus vaccines, the transgene antigen remains available to the system for a longer period than would be expected, resulting in a T-cell population with a sustained effector phenotype. The implications of these observations with regards to the utility of adenovirus vaccines are discussed.
Collapse
Affiliation(s)
- Teng Chih Yang
- Center for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada.
| | | | | | | | | | | |
Collapse
|
29
|
Liu W, Evanoff DP, Chen X, Luo Y. Urinary bladder epithelium antigen induces CD8+ T cell tolerance, activation, and autoimmune response. THE JOURNAL OF IMMUNOLOGY 2007; 178:539-46. [PMID: 17182594 PMCID: PMC4596412 DOI: 10.4049/jimmunol.178.1.539] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The effort to explore the specific autoimmune mechanisms of urinary bladder has long been hindered due to a lack of proper animal models. To better elucidate this issue, we developed a novel line of transgenic (Tg) mice, designated as URO-OVA mice, that express the model Ag OVA as a "self"-Ag on the bladder epithelium. URO-OVA mice are naturally tolerant to OVA and show no response to OVA stimulation. Adoptive transfer of naive OVA-specific T cells showed cell proliferation, activation, and infiltration but no bladder histopathology. In contrast, adoptive transfer of activated OVA-specific T cells induced OVA-mediated histological bladder inflammation. Increased mast cells and up-regulated mRNA expressions of TNF-alpha, nerve growth factor, and substance P precursor were also observed in the inflamed bladder. To further facilitate bladder autoimmunity study, we crossbred URO-OVA mice with OVA-specific CD8(+) TCR Tg mice (OT-I mice) to generate a dual Tg line URO-OVA/OT-I mice. The latter mice naturally acquire clonal deletion for autoreactive OT-I CD8(+) T cells (partial deletion in the thymus and severe deletion in the periphery). Despite this clonal deletion, URO-OVA/OT-I mice spontaneously develop autoimmune cystitis at 10 wk of age. Further studies demonstrated that the inflamed bladder contained infiltrating OT-I CD8(+) T cells that had escaped clonal deletion and gained effector functions before developing histological bladder inflammation. Taken together, we demonstrate for the first time that the bladder epithelium actively presents self-Ag to the immune system and induces CD8(+) T cell tolerance, activation, and autoimmune response.
Collapse
Affiliation(s)
| | | | | | - Yi Luo
- Address correspondence and reprint requests to Dr. Yi Luo, Department of Urology, University of Iowa, 3202 Medical Education and Research Facility, 375 Newton Road, Iowa City, IA 52242-1087.
| |
Collapse
|
30
|
Lees JR, Charbonneau B, Swanson AK, Jensen R, Zhang J, Matusik R, Ratliff TL. Deletion is neither sufficient nor necessary for the induction of peripheral tolerance in mature CD8+ T cells. Immunology 2006; 117:248-61. [PMID: 16423061 PMCID: PMC1782220 DOI: 10.1111/j.1365-2567.2005.02293.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Previous reports have demonstrated clonal deletion of CD8(+) T cells during peripheral tolerance induction to tissue antigens. However, direct evidence demonstrating a causal connection between deletion and tolerance has not been reported because of model limitations in which the tissue antigens were expressed in vital organs. Thus, studies were initiated in a mouse model where expression of a membrane-bound ovalbumin fusion protein (mOVA) was driven by a prostate specific androgen regulated probasin promotor, providing restricted expression in a non-vital organ where antigen levels can be abrogated through androgen deprivation. Adoptive transfer of mOVA specific CD8(+) T cells (OT-I) was used to assess the development of peripheral tolerance. Proliferation of OT-I cells was observed, as was partial deletion of transferred OT-I cells. Although deletion occurred, the long-term persistence of a stable level of OT-I cells was observed. Importantly, the persistent OT-I cells lost antigen responsiveness within 3 weeks of transfer. Castration resulted in loss of high-level prostate mOVA expression, with a resultant abrogation of tolerance induction, but surprisingly did not affect the deletion rate of OT-I cells. In contrast, abrogation of deletion through the adoptive transfer of OT-I cells from third generation CD95-deficient mice had no effect on tolerance induction. These data demonstrate the necessity for continued expression of tissue antigen throughout the establishment of peripheral tolerance. Furthermore, these findings demonstrate that deletion is neither sufficient nor required for CD8(+) T-cell tolerance to tissue antigens, suggesting that regulatory events independent of deletion are necessary for peripheral tolerance induction to prostate antigens.
Collapse
Affiliation(s)
- Jason R Lees
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, 52242, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Lubaroff DM, Konety B, Link BK, Ratliff TL, Madsen T, Shannon M, Ecklund D, Williams RD. Clinical protocol: phase I study of an adenovirus/prostate-specific antigen vaccine in men with metastatic prostate cancer. Hum Gene Ther 2006; 17:220-9. [PMID: 16454655 DOI: 10.1089/hum.2006.17.220] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- David M Lubaroff
- Department of Urology and Roland and Ruby Holden Cancer Research Laboratories, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Advances in gene modification and viral therapy have led to the development of a variety of vectors in several viral families that are capable of replication specifically in tumor cells. Because of the nature of viral delivery, infection, and replication, this technology, oncolytic virotherapy, may prove valuable for treating cancer patients, especially those with inoperable tumors. Current limitations exist, however, for oncolytic virotherapy. They include the body's B and T cell responses, innate inflammatory reactions, host range, safety risks involved in using modified viruses as treatments, and the requirement that most currently available oncolytic viruses require local administration. Another important constraint is that genetically enhanced vectors may or may not adhere to their replication restrictions in long-term applications. Several solutions and strategies already exist, however, to minimize or circumvent many of these limitations, supporting viral oncolytic therapy as a viable option and powerful tool in the fight against cancer.
Collapse
Affiliation(s)
- J J Davis
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
33
|
Elzey BD, Sprague DL, Ratliff TL. The emerging role of platelets in adaptive immunity. Cell Immunol 2006; 238:1-9. [PMID: 16442516 DOI: 10.1016/j.cellimm.2005.12.005] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2005] [Revised: 12/10/2005] [Accepted: 12/13/2005] [Indexed: 12/22/2022]
Abstract
Platelets' foremost role in survival is hemostasis. However, a significant quantity of research has demonstrated that platelets are an integral part of inflammation and can also be potent effector cells of the innate immune response. CD154, a molecule of vital importance to adaptive immune responses, is expressed by activated platelets and has been implicated in platelet-mediated modulation of innate immunity and inflammatory disease states. Recent studies in mice extend the role of platelet CD154 to the adaptive immune response demonstrating that platelets can enhance antigen presentation, improve CD8 T cell responses, and play a critical function in normal T-dependent humoral immunity. The latter studies suggest that the current paradigm for the B cell germinal center response should be modified to include a role for platelets.
Collapse
|
34
|
Lubaroff DM, Konety B, Link BK, Link TL, Madsen T, Shannon M, Ecklund D, Williams RD. Clinical Protocol: Phase I Study of an Adenovirus/Prostate-Specific Antigen Vaccine in Men with Metastatic Prostate Cancer. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
35
|
Irie A, Matsumoto K, Anderegg B, Kuruma H, Kashani-Sabet M, Scanlon KJ, Uchida T, Baba S. Growth inhibition efficacy of an adenovirus expressing dual therapeutic genes, wild-type p53, and anti-erbB2 ribozyme, against human bladder cancer cells. Cancer Gene Ther 2005; 13:298-305. [PMID: 16110311 DOI: 10.1038/sj.cgt.7700892] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The altered expression of both p53 and erbB2 is strongly related to the disease status and the outcome of bladder cancers. We examined the antitumor efficacy by the modulation of these genetic alterations with a newly designed dual-gene-expressing adenovirus (Ad-p53/erbB2Rz), which expresses p53 and anti-erbB2 ribozyme simultaneously in human bladder cancer cells. Cell growth inhibition efficacy along with biological responses of this virus was compared with other viral vectors (Ad-p53, which expresses wild-type p53 cDNA, and Ad-erbB2Rz, which expresses anti-erbB2 ribozyme, solely or in combination). Sufficient transgene expression in targeted cells and the altered expression of the targeted genes and their encoded proteins were obtained by each therapeutic vector. Each of the three therapeutic viral vectors inhibited bladder cancer cell growth, and the putative additive antitumor effect was shown by the combination of two of the therapeutic vectors. Furthermore, Ad-p53/erbB2Rz had superior therapeutic efficacy when the same titers of viruses were infected. Nonspecific vector-related toxicity was minimized by reducing the total amount of viral titers by using the dual-gene-expressing adenovirus. Modulation of multiple genetic abnormalities might enhance the therapeutic efficacy, and vector-related toxicity could be minimized when the total amount of viral titers are reduced.
Collapse
Affiliation(s)
- A Irie
- Department of Urology, Kitasato University School of Medicine, Kanagawa, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Jacob D, Davis JJ, Zhang L, Zhu H, Teraishi F, Fang B. Suppression of pancreatic tumor growth in the liver by systemic administration of the TRAIL gene driven by the hTERT promoter. Cancer Gene Ther 2005; 12:109-15. [PMID: 15486557 DOI: 10.1038/sj.cgt.7700773] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Local and locoregional administration of adenovectors expressing the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene has been demonstrated to be useful in treating established tumors in animals. Moreover, expression of the TRAIL gene from the human telomerase reverse transcriptase (hTERT) promoter can be used to prevent possible liver toxicity of the TRAIL gene. However, it remains unknown whether systemic administration of the TRAIL-expressing adenovector can be used for cancer therapy. Here, we showed that a combination of TRAIL gene therapy and gemcitabine, the first-line chemotheraphy agent for pancreatic cancer, had a synergistic effect on the induction of apoptosis in human pancreatic cancer cell lines in vitro. Systemic administration of an adenovector that contains an insertion of integrin-binding motif argine-glycine-aspartate (RGD) in the HI loop of the adenoviral fiber protein and expresses the human TRAIL gene from the hTERT promoter (designated Ad/TRAIL-F/RGD) suppressed the growth of human pancreatic tumor cells inoculated in the liver of nu/nu nude mice. Furthermore, Ad/TRAIL-F/RGD in combination with gemcitabine suppressed the tumor growth of pancreatic cancer in the liver more than did treatments consisting of each agent alone. No obvious liver toxicity was detected in any of the treatment groups. Our results suggest that TRAIL gene therapy in combination with gemcitabine might be a useful therapeutic approach for treating metastatic pancreatic cancers.
Collapse
Affiliation(s)
- Dietmar Jacob
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
37
|
Tscharke DC, Suhrbier A. From mice to humans – murine intelligence for human CD8+T cell vaccine design. Expert Opin Biol Ther 2005; 5:263-71. [PMID: 15757387 DOI: 10.1517/14712598.5.2.263] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There has been considerable progress in the design of vaccines capable of safely and effectively inducing CD8(+) T cells for prophylaxis and treatment of chronic infectious diseases and cancer. Much of what is known about CD8(+) T cell-mediated immunity has come from pioneering work in mice; this broad overview discusses recent work in mouse systems where lessons may be drawn for human vaccine development. The areas highlighted include antivector immunity, immunodominance, dendritic cell biology and targeting, the role of Toll-like receptors and their exploitation by novel adjuvants, the role of CD4(+) T cell help, regulatory T cells and, finally, some comments on the different requirements of prophylactic versus therapeutic vaccines.
Collapse
Affiliation(s)
- David C Tscharke
- Queensland Institute of Medical Research, EBVBiology, P.O. Royal Brisbane Hospital, Brisbane, QLD 4029, Australia.
| | | |
Collapse
|
38
|
Harui A, Roth MD, Kiertscher SM, Mitani K, Basak SK. Vaccination with helper-dependent adenovirus enhances the generation of transgene-specific CTL. Gene Ther 2005; 11:1617-26. [PMID: 15295617 DOI: 10.1038/sj.gt.3302332] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant adenoviral vectors (AdV) have been used experimentally as vaccines to present antigenic transgenes in vivo. However, administration of first-generation vectors (FG-AdV) is often limited by their induction of antiviral immunity. To address this limitation, helper-dependent vectors (HD-AdV) were developed that lack viral coding regions. While the administration of HD-AdV results in long-term gene expression in vivo, their utility as immunogens has never been examined. Direct vaccination with 10(8) blue-forming units (BFU) of HD-AdV injected into C57BL/6 mice lead to superior transgene-specific CTL and antibody responses when compared to the same amount of a FG-AdV. The antibody responses to viral antigens were high in response to both the vectors. As a mechanism to reduce viral exposure, dendritic cells (DC) were transduced with HD-AdV in vitro and then used as a cell-based vaccine. DC transduced with HD-AdV expressed higher levels of transgene-specific mRNA and up to 1200-fold higher levels of transgene protein than did DC transduced with a FG-AdV. In addition, HD-AdV-transduced DC stimulated superior transgene-specific CTL responses when administered in vivo, an effect that was further enhanced by maturing the DC with LPS prior to administration. In contrast to direct immunization with HD-AdV, vaccination with HD-AdV-transduced DC was associated with limited antibody responses against the AdV. We conclude that HD-AdV stimulates superior transgene-specific immune responses when compared to a FG-AdV, and that immunization with a DC-based vaccine maintains this efficacy while limiting antiviral reactivity.
Collapse
Affiliation(s)
- A Harui
- Pulmonary & Critical Care Medicine, UCLA School of Medicine, Los Angeles, CA 90095-1690, USA
| | | | | | | | | |
Collapse
|
39
|
Lin T, Gu J, Zhang L, Davis JJ, Huang X, Cabbini G, Ji L, Fang B. Enhancing adenovirus-mediated gene transfer in vitro and in vivo by addition of protamine and hydrocortisone. J Gene Med 2004; 5:868-875. [PMID: 14533195 DOI: 10.1002/jgm.427] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Inclusion of positively charged polymers such as protamine in adenovector formulations has been reported to improve the efficiency of adenovirus-mediated gene transfer in vitro and in vivo. On the other hand, corticosteroids are known to inhibit inflammation and thus might be useful in minimizing vector-related toxicity. In this study, we evaluated the combined effect of protamine sulfate and hydrocortisone on the efficiency of adenovirus-mediated gene transfer in vitro and in vivo. METHODS Protamine and hydrocortisone at different concentrations were added to adenovector formulations. In vitro transgene expression with or without inclusion of protamine and hydrocortisone was evaluated in the breast cancer cell lines MDA-MB-231 and MCF7 and the lung cancer cell lines A549 and H460. In vivo transgene expression in the mouse lung was determined after aerosolized vector delivery. RESULTS The combination of 2 micro g/ml protamine and 125 ng/ml hydrocortisone significantly increased transgene expression in vitro in all the cell lines tested. Protamine is only effective when it is added to cells before or together with adenovectors, whereas hydrocortisone is effective when it is added to cells before, together with, or after adenovectors. Inclusion of protamine and hydrocortisone also augmented apoptosis induction caused by adenovectors expressing proapoptotic genes in cancer cells. Moreover, protamine and hydrocortisone dramatically enhanced transgene expression in the mouse lung after aerosolized vector delivery. CONCLUSIONS Inclusion of protamine and hydrocortisone in adenovector formulations can improve adenovector-mediated gene expression and may be useful for clinical applications of current adenovirus-mediated gene therapy.
Collapse
Affiliation(s)
- Tongyu Lin
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Gu
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Lidong Zhang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - John J Davis
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
- The Program in Virology and Gene Therapy, The University of Texas Graduate School of Biomedical Sciences at The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Xuefeng Huang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Giovanni Cabbini
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Lin Ji
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
- The Program in Virology and Gene Therapy, The University of Texas Graduate School of Biomedical Sciences at The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
40
|
Basak SK, Kiertscher SM, Harui A, Roth MD. Modifying Adenoviral Vectors for Use as Gene-Based Cancer Vaccines. Viral Immunol 2004; 17:182-96. [PMID: 15279698 DOI: 10.1089/0882824041310603] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The past decade has produced significant advances in our understanding of antigen-presenting cells, tumor antigens, and other components of the immune response to cancer. Gene-based vaccination is emerging as one of the more promising approaches for loading dendritic cells (DC) with tumor-associated antigens. In this respect, it is proposed that adenoviral (AdV) vectors can deliver high antigen concentrations, promote effective processing and MHC expression, and stimulate potent cell-mediated immunity. While AdV vectors have performed well in pre-clinical vaccine models, their application to patient care has limitations. The in vivo administration of AdV vectors is associated with both innate and adaptive host responses that result in tissue inflammation and injury, viral neutralization, and premature clearance of AdV-transduced cells. A variety of strategies have been developed to address these limitations. The ideal vaccine would avoid vector-related immune responses, have relative specificity for transducing DC, and induce high levels of transgene expression. This review describes the range of host responses to AdV vaccines, identifies strategies to reduce viral recognition and enhance transgene antigen expression, and suggests future approaches to vector development and administration. There is every reason to believe that safer and more effective forms of AdV-based vaccines can be developed and applied to patient therapy.
Collapse
Affiliation(s)
- Saroj K Basak
- Pulmonary and Critical Care Medicine and the Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1690, USA
| | | | | | | |
Collapse
|
41
|
Elzey BD, Tian J, Jensen RJ, Swanson AK, Lees JR, Lentz SR, Stein CS, Nieswandt B, Wang Y, Davidson BL, Ratliff TL. Platelet-mediated modulation of adaptive immunity. A communication link between innate and adaptive immune compartments. Immunity 2003; 19:9-19. [PMID: 12871635 DOI: 10.1016/s1074-7613(03)00177-8] [Citation(s) in RCA: 291] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelets are highly reactive components of the circulatory system with well-documented hemostatic function. Recent studies extend platelet function to modulation of local inflammatory events through the release of chemokines, cytokines, and a number of immunomodulatory ligands, including CD154. We hypothesized that platelet-derived CD154 modulates adaptive immunity. The data reported herein demonstrate that platelets, via CD154, induce dendritic cell maturation, B cell isotype switching, and augment CD8(+) T cell responses both in vitro and in vivo. Platelet transfusion studies demonstrate that platelet-derived CD154 alone is sufficient to induce isotype switching and augment T lymphocyte function during viral infection, leading to enhanced protection against viral rechallenge. Additionally, depletion of platelets in normal mice results in decreased antigen-specific antibody production.
Collapse
Affiliation(s)
- Bennett D Elzey
- The University of Iowa, Department of Urology, Iowa City, IA 52242, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Siemens DR, Ratliff TL. Are Vaccinations for Prostate Cancer Realistic? Prostate Cancer 2003. [DOI: 10.1016/b978-012286981-5/50060-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
43
|
Huang X, Lin T, Gu J, Zhang L, Roth JA, Stephens LC, Yu Y, Liu J, Fang B. Combined TRAIL and Bax gene therapy prolonged survival in mice with ovarian cancer xenograft. Gene Ther 2002; 9:1379-86. [PMID: 12365003 DOI: 10.1038/sj.gt.3301810] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2002] [Accepted: 05/07/2002] [Indexed: 11/08/2022]
Abstract
We evaluated the antitumor activity of the Bax gene and green fluorescent protein/tumor necrosis factor-related apoptosis-inducing ligand (GFP/TRAIL) fusion gene driven by the human telomerase reverse transcriptase promoter both separately and combined in the human ovarian cancer lines SKOV3ip and DOV13 and human lung cancer line H1299. In vitro study showed that both TRAIL- and Bax-expressing vectors elicited significant cell killing in H1299 and SKOV3ip cells, but only the GFP/TRAIL gene elicited significant cell killing in DOV13 cells. Combined TRAIL and Bax therapy also produced more profound cell killing in SKOV3ip and H1299 cells, but not DOV13 cells without escalation of the vector doses. To further evaluate the combined effects of Bax and TRAIL, abdominally spread tumors were established in nude mice via intraperitoneal inoculation of SKOV3ip cells followed by that of adenoviral vectors. Tumor growth, ascites formation, survival duration and toxicity were evaluated after treatment. We found that treatment using the Bax- or TRAIL-expressing vector alone significantly suppressed tumor growth and ascites formation, and prolonged animal survival when compared with that of using PBS or a control vector. Combined TRAIL and Bax therapy further prolonged survival significantly when compared with therapy using the TRAIL or Bax gene alone. Transgene expression and apoptosis induction were not detected in normal human ovarian epithelial cells in vitro or normal mouse tissues in vivo after intraperitoneal vector administration. Also, liver toxicity was not detected after either treatment. Thus, combined TRAIL and Bax gene therapy may be useful for treatment of abdominally spread tumors.
Collapse
Affiliation(s)
- X Huang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Griffith TS, Broghammer EL. Suppression of tumor growth following intralesional therapy with TRAIL recombinant adenovirus. Mol Ther 2001; 4:257-66. [PMID: 11545617 DOI: 10.1006/mthe.2001.0439] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
TRAIL is a member of the tumor necrosis factor superfamily that induces apoptosis in a variety of tumor cell types both in vitro and in vivo, while demonstrating minimal cytotoxicity toward normal tissues. One disadvantage to previous in vivo protocols was the need for large quantities of TRAIL to suppress tumor growth. Here we engineered a replication-deficient adenovirus to encode human TNFSF10 (Ad5-TRAIL) as an alternative to recombinant, soluble TRAIL protein. The results show that TRAIL-sensitive prostate tumor cell targets infected with Ad5-TRAIL undergo apoptosis through the production and expression of TRAIL protein. This activity was limited to TRAIL-sensitive tumor cells, as normal prostate epithelial cells were not killed by Ad5-TRAIL. Furthermore, in vivo administration of Ad5-TRAIL at the site of tumor implantation suppressed the outgrowth of human prostate tumor xenografts in SCID mice. Histologic examination of prostate tumors treated locally with Ad5-TRAIL revealed areas of apoptosis within 24 hours of injection. These results further define Ad5-TRAIL as a novel anti-tumor therapeutic and demonstrate its potential use as a means for treating prostate tumors, as well as other solid tumors, in vivo.
Collapse
Affiliation(s)
- T S Griffith
- Department of Urology, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242-1089, USA.
| | | |
Collapse
|
45
|
Steitz J, Brück J, Knop J, Tüting T. Adenovirus-transduced dendritic cells stimulate cellular immunity to melanoma via a CD4(+) T cell-dependent mechanism. Gene Ther 2001; 8:1255-63. [PMID: 11509959 DOI: 10.1038/sj.gt.3301521] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2001] [Accepted: 05/23/2001] [Indexed: 11/08/2022]
Abstract
We previously showed that genetic immunization of C57BL/6 mice with recombinant adenovirus encoding human TRP2 (Ad-hTRP2) was able to circumvent tolerance and induce cellular and humoral immune responses to murine TRP2 associated with protection against metastatic growth of B16 melanoma. In the present study we compared delivery of Ad-hTRP2 with cultured dendritic cells (DC) and direct injections of Ad-hTRP2. We show that application of Ad-hTRP2 with cultured DC enhanced protective immunity to B16 melanoma cells. Most importantly, delivery of recombinant adenovirus with DC alters the character of the immune response resulting in preferential stimulation of strong cellular immunity in the absence of significant humoral immunity to the encoded antigen. Adoptive transfer of lymphocytes from mice immunized with Ad-hTRP2-transduced DC confirmed that cellular components of the immune response were responsible for rejection of B16 melanoma. The protective efficacy of Ad-hTRP2-transduced DC clearly depended on the presence of CD4(+) T helper cells. Furthermore, AD-hTRP2-transduced DC, but not direct injection of Ad-hTRP2, were effective in the presence of neutralizing anti-adenoviral antibodies. These preclinical studies demonstrate the superiority of melanoma vaccines consisting of cultured DC transduced with recombinant adenoviruses encoding melanoma antigens.
Collapse
Affiliation(s)
- J Steitz
- Department of Dermatology, J Gutenberg-University, Langenbeckstr 1, D-55101 Mainz, Germany
| | | | | | | |
Collapse
|