1
|
Hasanpourghadi M, Novikov M, Ambrose R, Chekaoui A, Newman D, Xiang Z, Luber AD, Currie SL, Zhou X, Ertl HC. A therapeutic HBV vaccine containing a checkpoint modifier enhances CD8+ T cell and antiviral responses. JCI Insight 2024; 9:e181067. [PMID: 39226106 PMCID: PMC11601613 DOI: 10.1172/jci.insight.181067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024] Open
Abstract
In patients who progress from acute hepatitis B virus (HBV) infection to a chronic HBV (CHB) infection, CD8+ T cells fail to eliminate the virus and become impaired. A functional cure of CHB likely requires CD8+ T cell responses different from those induced by the infection. Here we report preclinical immunogenicity and efficacy of an HBV therapeutic vaccine that includes herpes simplex virus (HSV) glycoprotein D (gD), a checkpoint modifier of early T cell activation, that augments CD8+ T cell responses. The vaccine is based on a chimpanzee adenovirus serotype 6 (AdC6) vector, called AdC6-gDHBV2, which targets conserved and highly immunogenic regions of the viral polymerase and core antigens fused to HSV gD. The vaccine was tested with and without gD in mice for immunogenicity, and in an AAV8-1.3HBV vector model of antiviral efficacy. The vaccine encoding the HBV antigens within gD stimulates potent and broad CD8+ T cell responses. In a surrogate model of HBV infection, a single intramuscular injection achieved pronounced and sustained declines of circulating HBV DNA copies and HBV surface antigen; both inversely correlated with HBV-specific CD8+ T cell frequencies in spleen and liver.
Collapse
Affiliation(s)
| | | | | | | | - Dakota Newman
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - ZhiQuan Xiang
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
2
|
Mohammadi M, Saha A, Giles-Davis W, Xiang Z, Novikov M, Hasanpourghadi M, C. J. Ertl H. Preclinical Immunogenicity and Efficacy Studies for Therapeutic Vaccines for Human Papillomavirus-Type-16-Associated Cancer. Vaccines (Basel) 2024; 12:616. [PMID: 38932345 PMCID: PMC11209626 DOI: 10.3390/vaccines12060616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The objective of this study was to conduct preclinical immunogenicity and efficacy studies with several therapeutic vaccines for human papillomavirus (HPV)-16-associated cancers expressing the early antigens E5, E6, and E7 with or without E2. The viral oncoproteins were either expressed by themselves as fusion proteins or the fusion proteins were inserted genetically into herpes simplex virus (HSV)-1 glycoprotein D (gD) which, upon binding to the herpes virus entry mediator (HVEM), inhibits an early T cell checkpoint mediated by the B and T cell mediator (BTLA). This, in turn, lowers the threshold for T cell activation and augments and broadens CD8+ T cell responses to the antigens. The fusion antigens were expressed by chimpanzee adenovirus (AdC) vectors. Expression of the HPV antigens within gD was essential for vaccine immunogenicity and efficacy against challenge with TC-1 cells, which express E7 and E6 of HPV-16 but neither E5 nor E2. Unexpectedly, inclusion of E2 increased both CD8+ T cell responses to the other oncoproteins of HPV-16 and the effectiveness of the vaccines to cause the regression of sizable TC-1 tumors.
Collapse
|
3
|
Hasanpourghadi M, Chekaoui A, Kurian S, Kurupati R, Ambrose R, Giles-Davis W, Saha A, Xiaowei X, Ertl HC. Treatment with the PPARα agonist fenofibrate improves the efficacy of CD8 + T cell therapy for melanoma. Mol Ther Oncolytics 2023; 31:100744. [PMID: 38075243 PMCID: PMC10701456 DOI: 10.1016/j.omto.2023.100744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/31/2023] [Indexed: 02/12/2024] Open
Abstract
Adoptive transfer of tumor antigen-specific CD8+ T cells can limit tumor progression but is hampered by the T cells' rapid functional impairment within the tumor microenvironment (TME). This is in part caused by metabolic stress due to lack of oxygen and glucose. Here, we report that fenofibrate treatment of human ex vivo expanded tumor-infiltrating lymphocytes (TILs) improves their ability to limit melanoma progression in a patient-derived xenograft (PDX) mouse model. TILs treated with fenofibrate, a peroxisome proliferator receptor alpha (PPARα) agonist, switch from glycolysis to fatty acid oxidation (FAO) and increase the ability to slow the progression of autologous melanomas in mice with freshly transplanted human tumor fragments or injected with tumor cell lines established from the patients' melanomas and ex vivo expanded TILs.
Collapse
Affiliation(s)
| | | | | | - Raj Kurupati
- The Wistar Institute, Philadelphia, PA 19104, USA
- The Janssen Pharmaceutical Companies of Johnson & Johnson, New Brunswick, NJ, USA
| | | | | | - Amara Saha
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Xu Xiaowei
- Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
4
|
Hasanpourghadi M, Novikov M, Ambrose R, Chekaoui A, Newman D, Ding J, Giles-Davis W, Xiang Z, Zhou XY, Liu Q, Swagata K, Ertl HCJ. Heterologous chimpanzee adenovirus vector immunizations for SARS-CoV-2 spike and nucleocapsid protect hamsters against COVID-19. Microbes Infect 2022; 25:105082. [PMID: 36539010 PMCID: PMC9758783 DOI: 10.1016/j.micinf.2022.105082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Available COVID-19 vaccine only provide protection for a limited time due in part to the rapid emergence of viral variants with spike protein mutations, necessitating the generation of new vaccines to combat SARS-CoV-2. Two serologically distinct replication-defective chimpanzee-origin adenovirus (Ad) vectors (AdC) called AdC6 and AdC7 expressing early SARS-CoV-2 isolate spike (S) or nucleocapsid (N) proteins, the latter expressed as a fusion protein within herpes simplex virus glycoprotein D (gD), were tested individually or as a mixture in a hamster COVID-19 SARS-CoV-2 challenge model. The S protein expressing AdC (AdC-S) vectors induced antibodies including those with neutralizing activity that in part cross-reacted with viral variants. Hamsters vaccinated with the AdC-S vectors were protected against serious disease and showed accelerated recovery upon SARS-CoV-2 challenge. Protection was enhanced if AdC-S vectors were given together with the AdC vaccines that expressed the gD N fusion protein (AdC-gDN). In contrast hamsters that just received the AdC-gDN vaccines showed only marginal lessening of symptoms compared to control animals. These results indicate that immune response to the N protein that is less variable than the S protein may potentiate and prolong protection achieved by the currently used S protein based genetic COVID-19 vaccines.
Collapse
Affiliation(s)
| | - Mikhail Novikov
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | - Robert Ambrose
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | - Arezki Chekaoui
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | - Dakota Newman
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | - Jianyi Ding
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | | | - Zhiquan Xiang
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | - Xiang Yang Zhou
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | - Qin Liu
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | - Kar Swagata
- Bioqual Inc., 9600 Medical Center Dr #101, Rockville, MD 20850
| | - Hildegund CJ. Ertl
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA,Corresponding author. Tel.: +1 215-898-3863
| |
Collapse
|
5
|
Hasanpourghadi M, Novikov M, Newman D, Xiang Z, Zhou XY, Magowan C, Ertl HCJ. Hepatitis B virus polymerase-specific T cell epitopes shift in a mouse model of chronic infection. Virol J 2021; 18:242. [PMID: 34876153 PMCID: PMC8650432 DOI: 10.1186/s12985-021-01712-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 11/26/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chronic hepatitis B virus (HBV) infection (CHB) is a significant public health problem that could benefit from treatment with immunomodulators. Here we describe a set of therapeutic HBV vaccines that target the internal viral proteins. METHODS Vaccines are delivered by chimpanzee adenovirus vectors (AdC) of serotype 6 (AdC6) and 7 (AdC7) used in prime only or prime-boost regimens. The HBV antigens are fused into an early T cell checkpoint inhibitor, herpes simplex virus (HSV) glycoprotein D (gD), which enhances and broadens vaccine-induced cluster of differentiation (CD8)+ T cell responses. RESULTS Our results show that the vaccines are immunogenic in mice. They induce potent CD8+ T cell responses that recognize multiple epitopes. CD8+ T cell responses increase after a boost, although the breadth remains similar. In mice, which carry high sustained loads of HBV particles due to a hepatic infection with an adeno-associated virus (AAV)8 vector expressing the 1.3HBV genome, CD8+ T cell responses to the vaccines are attenuated with a marked shift in the CD8+ T cells' epitope recognition profile. CONCLUSIONS Our data show that in different stains of mice including those that carry a human major histocompatibility complex (MHC) class I antigen HBV vaccines adjuvanted with a checkpoint inhibitor induce potent and broad HBV-specific CD8+ T cell responses and lower but still detectable CD4+ T cell responses. CD8+ T cell responses are reduced and their epitope specificity changes in mice that are chronically exposed to HBV antigens. Implications for the design of therapeutic HBV vaccines are discussed.
Collapse
Affiliation(s)
| | - Mikhail Novikov
- Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Dakota Newman
- Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - ZhiQuan Xiang
- Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Xiang Yang Zhou
- Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Colin Magowan
- Virion Therapeutics LLC, 7 Creek Bend Ct, Newark, DE, 19711, USA
| | | |
Collapse
|
6
|
Chekaoui A, Ertl HCJ. PPARα Agonist Fenofibrate Enhances Cancer Vaccine Efficacy. Cancer Res 2021; 81:4431-4440. [PMID: 34244236 DOI: 10.1158/0008-5472.can-21-0052] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/27/2021] [Accepted: 07/07/2021] [Indexed: 11/16/2022]
Abstract
Reducing metabolic stress within the tumor microenvironment (TME) could be essential for improving the efficacy of cancer immunotherapy. Using a mouse model of melanoma, we show here that appropriately timed treatment with the PPARα agonist fenofibrate improves the ability of a T cell-inducing cancer vaccine to delay tumor progression. Fenofibrate reduced the use of glucose by tumor and stromal cells in the TME and promoted the use of fatty acids for their metabolic needs. The glucose within the TME was in turn available for use by vaccine-induced tumor-infiltrating CD8+ T cells, which improved their ability to slow tumor progression. Early fenofibrate treatment 3 days after vaccination improved functions of circulating CD8+ T cells but failed to significantly affect tumor-infiltrating lymphocyte (TIL) metabolism or decrease tumor progression. In contrast, delaying treatment until day 5 after vaccination modified TIL metabolism and augmented the vaccine's ability to slow tumor progression. In summary, our findings reveal that a PPARα agonist can increase the efficacy of a cancer vaccine by reprogramming cells within tumors to increase fatty acid metabolism, providing T cells access to glucose in the TME. SIGNIFICANCE: These findings suggest that metabolic manipulations using already approved drugs may offer an easy pathway to increase the efficacy of vaccines against solid tumors.
Collapse
|
7
|
Hasanpourghadi M, Novikov M, Ambrose R, Chekaoui A, Newman D, Zhou XY, Ertl HCJ. T cell responses to adenoviral vectors expressing the SARS-CoV-2 nucleoprotein. CURRENT TRENDS IN MICROBIOLOGY 2021; 15:1-28. [PMID: 35903088 PMCID: PMC9328080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
SARS-CoV-2 vaccines aim to protect against COVID-19 through neutralizing antibodies against the viral spike protein. Mutations within the spike's receptor-binding domain may eventually reduce vaccine efficacy, necessitating periodic updates. Vaccine-induced immunity could be broadened by adding T cell-inducing antigens such as SARS-CoV-2's nucleoprotein (N). Here we describe two replication-defective chimpanzee adenovirus (AdC) vectors from different serotypes expressing SARS-CoV-2 N either in its wild-type form or fused into herpes simplex virus glycoprotein D (gD), an inhibitor of an early T cell checkpoint. The vaccines induce potent and sustained CD8+ T cell responses that are broadened upon inclusion of gD. Depending on the vaccine regimen booster immunizations increase magnitude and breadth of T cell responses. Epitopes that are recognized by the vaccine-induced T cells are highly conserved among global SARS-CoV-2 isolates indicating that addition of N to COVID-19 vaccines may lessen the risk of loss of vaccine-induced protection due to variants.
Collapse
Affiliation(s)
| | - Mikhail Novikov
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Robert Ambrose
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Arezki Chekaoui
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Dakota Newman
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Xiang Yang Zhou
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Hildegund C. J. Ertl
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| |
Collapse
|
8
|
Gmyrek GB, Filiberti A, Montgomery M, Chitrakar A, Royer DJ, Carr DJJ. Herpes Simplex Virus 1 (HSV-1) 0ΔNLS Live-Attenuated Vaccine Protects against Ocular HSV-1 Infection in the Absence of Neutralizing Antibody in HSV-1 gB T Cell Receptor-Specific Transgenic Mice. J Virol 2020; 94:e01000-20. [PMID: 32999018 PMCID: PMC7925190 DOI: 10.1128/jvi.01000-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022] Open
Abstract
The contribution of T cell and antibody responses following vaccination in resistance to herpes simplex virus 1 (HSV-1) infection continues to be rigorously investigated. In the present article, we explore the contribution of CD8+ T cells specific for the major antigenic epitope for HSV-1 glycoprotein B (gB498-505, gB) in C57BL/6 mice using a transgenic mouse (gBT-I.1) model vaccinated with HSV-1 0ΔNLS. gBT-I.1-vaccinated mice did not generate a robust neutralization antibody titer in comparison to the HSV-1 0ΔNLS-vaccinated wild-type C57BL/6 counterpart. Nevertheless, the vaccinated gBT-I.1 mice were resistant to ocular challenge with HSV-1 compared to vehicle-vaccinated animals based on survival and reduced corneal neovascularization but displayed similar levels of corneal opacity. Whereas there was no difference in the virus titer recovered from the cornea comparing vaccinated mice, HSV-1 0ΔNLS-vaccinated animals possessed significantly less infectious virus during acute infection in the trigeminal ganglia (TG) and brain stem compared to the control-vaccinated group. These results correlated with a significant increase in gB-elicited interferon-γ (IFN-γ), granzyme B, and CD107a and a reduction in lymphocyte activation gene 3 (LAG-3), programmed cell death 1 (PD-1), and T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) expressed by TG infiltrating gB-specific CD8+ T cells from the HSV-1 0ΔNLS-vaccinated group. Antibody depletion of CD8+ T cells in HSV-1 0ΔNLS-vaccinated mice rendered animals highly susceptible to virus-mediated mortality similar to control-vaccinated mice. Collectively, the HSV-1 0ΔNLS vaccine is effective against ocular HSV-1 challenge, reducing ocular neovascularization and suppressing peripheral nerve virus replication in the near absence of neutralizing antibody in this unique mouse model.IMPORTANCE The role of CD8+ T cells in antiviral efficacy using a live-attenuated virus as the vaccine is complicated by the humoral immune response. In the case of the herpes simplex virus 1 (HSV-1) 0ΔNLS vaccine, the correlate of protection has been defined to be primarily antibody driven. The current study shows that in the near absence of anti-HSV-1 antibody, vaccinated mice are protected from subsequent challenge with wild-type HSV-1 as measured by survival. The efficacy is lost following depletion of CD8+ T cells. Whereas increased survival and reduction in virus replication were observed in vaccinated mice challenged with HSV-1, cornea pathology was mixed with a reduction in neovascularization but no change in opacity. Collectively, the study suggests CD8+ T cells significantly contribute to the host adaptive immune response to HSV-1 challenge following vaccination with an attenuated virus, but multiple factors are involved in cornea pathology in response to ocular virus challenge.
Collapse
Affiliation(s)
- Grzegorz B Gmyrek
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Adrian Filiberti
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Micaela Montgomery
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Alisha Chitrakar
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Derek J Royer
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Daniel J J Carr
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
9
|
Kurupati RK, Zhou X, Xiang Z, Keller LH, Ertl HCJ. Safety and immunogenicity of a potential checkpoint blockade vaccine for canine melanoma. Cancer Immunol Immunother 2018; 67:1533-1544. [PMID: 30051333 PMCID: PMC7080056 DOI: 10.1007/s00262-018-2201-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 07/06/2018] [Indexed: 12/13/2022]
Abstract
Human immunotherapy with checkpoint blockades has achieved significant breakthroughs in recent years. In this study, a checkpoint blockade vaccine for canine melanoma was tested for safety and immunogenicity. Five healthy adult dogs received a mixture of three replication-defective chimpanzee-derived adenoviral vectors, one expressing mouse fibroblast-associated protein (mFAP) and the others expressing canine melanoma-associated antigens Trp-1 or Trp-2 fused into Herpes Simplex-1 glycoprotein D, a checkpoint inhibitor of herpes virus entry mediator (HVEM) pathways. The vaccine mixture was shown to be well tolerated and increased frequencies of canineTrp-1-specific activated CD8+ and CD4+ T cells secreting interferon-(IFN)-γ, tumor necrosis factor (TNF)-α, or interleukin (IL)-2 alone or in combinations in four and five out of five dogs, respectively. To avoid excessive bleeds, responses to cTrp-2 were not analyzed. All dogs responded with increased frequencies of mFAP-specific activated CD8+ and CD4+ T cells. The results of this safety/immunogenicity trial invite further testing of this checkpoint blockade vaccine combination in dogs with melanoma.
Collapse
Affiliation(s)
- Raj K Kurupati
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Xiangyang Zhou
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Zhiquan Xiang
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | | | | |
Collapse
|
10
|
Zhao H, Xu C, Luo X, Wei F, Wang N, Shi H, Ren X. Seroprevalence of Neutralizing Antibodies against Human Adenovirus Type-5 and Chimpanzee Adenovirus Type-68 in Cancer Patients. Front Immunol 2018; 9:335. [PMID: 29563911 PMCID: PMC5845880 DOI: 10.3389/fimmu.2018.00335] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/06/2018] [Indexed: 01/16/2023] Open
Abstract
Since the preclinical results about chimpanzee adenovirus serotype-68 (AdC68)-based vaccine showed an encouraging results, it reminded us that AdC68 may be a suitable cancer vaccine vector. Previous study indicated that the seroprevalence of neutralizing antibodies (NAbs) against adenovirus was different between cancer patients and healthy volunteers. Knowledge regarding the prevalence rates of AdC68 NAbs for cancer patients is lacking. Therefore, assessing the preexistence of NAbs against AdC68 in cancer patients could provide useful insights for developing future AdC68-based cancer vaccines. In this study, 440 patients with different pathological types of tumors and 204 healthy adult volunteers were enrolled to evaluate the NAbs against AdC68 and human adenovirus serotype-5 (AdHu5). The seroprevalence of NAbs against AdC68 was much lower than that against AdHu5 in cancer subjects (43.64 vs. 67.05%, P < 0.01). The seroprevalence rates of NAbs to AdC68 in the cancer subjects were statistically higher than those detected in the healthy adult volunteers (43.64 vs. 23.53%, P = 0.000). The seroprevalence rates of AdC68 NAbs were much lower in lung, laryngeal, esophageal, and cervical cancer patients compared with oropharyngeal, colon, and rectal cancer patients. Furthermore, the seroprevalence rates of AdC68 NAbs were much lower in lung adenocarcinoma patients than in lung squamous cell carcinoma patients (35.00 vs. 70.00%, P < 0.05). No significant difference in the AdC68 NAbs among patients with different clinical stages of cancer was detected. The percentage of NAbs against AdC68 was significantly lower than that against AdHu5 (P < 0.05) in stage-I, -II, and -III cancer patients. No significant difference between the percentage of NAbs against AdC68 and AdHu5 in the subjects with stage-IV cancer was detected. The study also demonstrated the distribution of AdHu5 and AdC68 NAb titers for the positive samples. It showed that very low NAb titers against AdC68 with respect to AdHu5 in both healthy subjects and cancer subjects, especially in lung, laryngeal, esophageal, gastric, and cervical carcinomas. Also, the titer of NAbs against AdC68 was significantly lower than that against AdHu5 in the same clinical stage and age group (P < 0.05). Taken together, the present study showed that NAbs against AdC68 is much lower than AdHu5, especially in lung adenocarcinoma, laryngeal cancer, esophageal cancer, and cervical cancer patients. These results provided strong support for candidating AdC68 as a suitable vector of cancer vaccines.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Can Xu
- Bioroc Pharmaceutical & Biotech Company, Tianjin, China.,Tianjin Genstar Vaccine Limited Liability Company, Tianjin, China
| | - Xiaoli Luo
- Bioroc Pharmaceutical & Biotech Company, Tianjin, China.,Tianjin Genstar Vaccine Limited Liability Company, Tianjin, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Ning Wang
- Bioroc Pharmaceutical & Biotech Company, Tianjin, China.,Tianjin Genstar Vaccine Limited Liability Company, Tianjin, China
| | - Huiying Shi
- Bioroc Pharmaceutical & Biotech Company, Tianjin, China.,Tianjin Genstar Vaccine Limited Liability Company, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| |
Collapse
|
11
|
de Annunzio SR, de Freitas LM, Blanco AL, da Costa MM, Carmona-Vargas CC, de Oliveira KT, Fontana CR. Susceptibility of Enterococcus faecalis and Propionibacterium acnes to antimicrobial photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 178:545-550. [PMID: 29253813 DOI: 10.1016/j.jphotobiol.2017.11.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/24/2017] [Accepted: 11/26/2017] [Indexed: 12/31/2022]
Abstract
Bacterial resistance to available antibiotics nowadays is a global threat leading researchers around the world to study new treatment modalities for infections. Antimicrobial photodynamic therapy (aPDT) has been considered an effective and promising therapeutic alternative in this scenario. Briefly, this therapy is based on the activation of a non-toxic photosensitizing agent, known as photosensitizer (PS), by light at a specific wavelength generating cytotoxic singlet oxygen and free radicals. Virtually all studies related to aPDT involve a huge screening to identify ideal PS concentration and light dose combinations, a laborious and time-consuming process that is hardly disclosed in the literature. Herein, we describe an antimicrobial Photodynamic Therapy (aPDT) study against Enterococcus faecalis and Propionibacterium acnes employing methylene blue, chlorin-e6 or curcumin as PS. Similarities and discrepancies between the two bacterial species were pointed out in an attempt to speed up and facilitate futures studies against those clinical relevant strains. Susceptibility tests were performed by the broth microdilution method. Our results demonstrate that aPDT mediated by the three above-mentioned PS was effective in eliminating both gram-positive bacteria, although P. acnes showed remarkably higher susceptibility to aPDT when compared to E. faecalis. PS uptake assays revealed that P. acnes is 80 times more efficient than E. faecalis in internalizing all three PS molecules. Our results evidence that the cell wall structure is not a limiting feature when predicting bacterial susceptibility to aPDT treatment.
Collapse
Affiliation(s)
- Sarah Raquel de Annunzio
- Universidade Estadual Paulista (Unesp), Faculdade de Ciências Farmacêuticas, Araraquara. Rodovia Araraquara-Jaú, Km1, Campus Ville, Araraquara, SP, CEP 14800-903, Brazil
| | - Laura Marise de Freitas
- Universidade Estadual Paulista (Unesp), Faculdade de Ciências Farmacêuticas, Araraquara. Rodovia Araraquara-Jaú, Km1, Campus Ville, Araraquara, SP, CEP 14800-903, Brazil
| | - Ana Lígia Blanco
- Universidade Estadual Paulista (Unesp), Faculdade de Ciências Farmacêuticas, Araraquara. Rodovia Araraquara-Jaú, Km1, Campus Ville, Araraquara, SP, CEP 14800-903, Brazil
| | - Mardoqueu Martins da Costa
- Universidade Brasil (UniBrasil), Departamento de Engenharia Biomédica, Rua Carolina Fonseca, 235, Vila Santana, São Paulo, SP CEP: 08230-030, Brazil.
| | - Christian C Carmona-Vargas
- Universidade Federal de São Carlos (UFSCar), Departamento de Química, Laboratório de Química Bioorgânica, Rodovia Washington Luis, Km 235 - SP-310, São Carlos, SP, CEP 13565-905, Brazil
| | - Kleber Thiago de Oliveira
- Universidade Federal de São Carlos (UFSCar), Departamento de Química, Laboratório de Química Bioorgânica, Rodovia Washington Luis, Km 235 - SP-310, São Carlos, SP, CEP 13565-905, Brazil.
| | - Carla Raquel Fontana
- Universidade Estadual Paulista (Unesp), Faculdade de Ciências Farmacêuticas, Araraquara. Rodovia Araraquara-Jaú, Km1, Campus Ville, Araraquara, SP, CEP 14800-903, Brazil.
| |
Collapse
|
12
|
Dawany N, Parzych EM, Showe LC, Ertl HC. Age-related changes in the gene expression profile of antigen-specific mouse CD8+ T cells can be partially reversed by blockade of the BTLA/CD160 pathways during vaccination. Aging (Albany NY) 2017; 8:3272-3297. [PMID: 27922818 PMCID: PMC5270668 DOI: 10.18632/aging.101105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/26/2016] [Indexed: 01/12/2023]
Abstract
We analyzed gene expression profiles of young and aged mouse CD8+ T cells specific for the nucleoprotein (NP) of influenza A/PR8/34 virus. CD8+ T cells were stimulated either by the NP antigen expressed in its native form or fused into the herpes virus (HSV)-1 glycoprotein D (gD) protein, which blocks signaling through the immunoinhibitory B and T lymphocyte attenuator (BTLA) and CD160 pathways. We show that NP-specific CD8+ T cells from aged mice exhibit numerous differences in gene expression compared to NP-specific CD8+ T cells from young mice, including a significant reduction of expression in genes involved in T cell receptor (TcR) and CD28 signaling. We also show that these changes can be reversed in a sub-population (∼50%) of the aged mice by a BTLA/CD160 checkpoint blockade. These results suggest that BTLA/CD160 checkpoint blockade has potential value as a vaccine additive to induce better CD8+ T cell responses in the aged.
Collapse
Affiliation(s)
- Noor Dawany
- Wistar Institute, Philadelphia, PA 19104, USA.,Present Address: The Children's Hospital of Philadelphia, Department of Biomedical and Health Informatics, Philadelphia, PA 19104, USA
| | - Elizabeth M Parzych
- Wistar Institute, Philadelphia, PA 19104, USA.,Present Address: Drexel University, School of Medicine, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
13
|
Zhang Y, Kurupati R, Liu L, Zhou XY, Zhang G, Hudaihed A, Filisio F, Giles-Davis W, Xu X, Karakousis GC, Schuchter LM, Xu W, Amaravadi R, Xiao M, Sadek N, Krepler C, Herlyn M, Freeman GJ, Rabinowitz JD, Ertl HCJ. Enhancing CD8 + T Cell Fatty Acid Catabolism within a Metabolically Challenging Tumor Microenvironment Increases the Efficacy of Melanoma Immunotherapy. Cancer Cell 2017; 32:377-391.e9. [PMID: 28898698 PMCID: PMC5751418 DOI: 10.1016/j.ccell.2017.08.004] [Citation(s) in RCA: 456] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 06/28/2017] [Accepted: 08/08/2017] [Indexed: 02/08/2023]
Abstract
How tumor-infiltrating T lymphocytes (TILs) adapt to the metabolic constrains within the tumor microenvironment (TME) and to what degree this affects their ability to combat tumor progression remain poorly understood. Using mouse melanoma models, we report that CD8+ TILs enhance peroxisome proliferator-activated receptor (PPAR)-α signaling and catabolism of fatty acids (FAs) when simultaneously subjected to hypoglycemia and hypoxia. This metabolic switch partially preserves CD8+ TILs' effector functions, although co-inhibitor expression increases during tumor progression regardless of CD8+ TILs' antigen specificity. Further promoting FA catabolism improves the CD8+ TILs' ability to slow tumor progression. PD-1 blockade delays tumor growth without changing TIL metabolism or functions. It synergizes with metabolic reprogramming of T cells to achieve superior antitumor efficacy and even complete cures.
Collapse
Affiliation(s)
- Ying Zhang
- Gene Therapy and Vaccines Program, University of Pennsylvania (U of PA), Philadelphia, PA 19104, USA; The Wistar Institute, Philadelphia, PA 19104, USA
| | - Raj Kurupati
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Ling Liu
- Lewis-Sigler Institute for Integrative Genomics & Department of Chemistry, Princeton University, Princeton, NJ 08540, USA
| | | | - Gao Zhang
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Abeer Hudaihed
- Biology Program, Temple University, Philadelphia, PA 19122, USA
| | - Flavia Filisio
- Biology Program, Temple University, Philadelphia, PA 19122, USA
| | | | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, U of PA, Philadelphia, PA 19104, USA
| | | | | | - Wei Xu
- Department of Medicine, U of PA, Philadelphia, PA 19104, USA
| | - Ravi Amaravadi
- Department of Medicine, U of PA, Philadelphia, PA 19104, USA
| | - Min Xiao
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Norah Sadek
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics & Department of Chemistry, Princeton University, Princeton, NJ 08540, USA
| | | |
Collapse
|
14
|
Is There Still Room for Cancer Vaccines at the Era of Checkpoint Inhibitors. Vaccines (Basel) 2016; 4:vaccines4040037. [PMID: 27827885 PMCID: PMC5192357 DOI: 10.3390/vaccines4040037] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/23/2016] [Accepted: 10/31/2016] [Indexed: 02/07/2023] Open
Abstract
Checkpoint inhibitor (CPI) blockade is considered to be a revolution in cancer therapy, although most patients (70%–80%) remain resistant to this therapy. It has been hypothesized that only tumors with high mutation rates generate a natural antitumor T cell response, which could be revigorated by this therapy. In patients with no pre-existing antitumor T cells, a vaccine-induced T cell response is a rational option to counteract clinical resistance. This hypothesis has been validated in preclinical models using various cancer vaccines combined with inhibitory pathway blockade (PD-1-PDL1-2, CTLA-4-CD80-CD86). Enhanced T cell infiltration of various tumors has been demonstrated following this combination therapy. The timing of this combination appears to be critical to the success of this therapy and multiple combinations of immunomodulating antibodies (CPI antagonists or costimulatory pathway agonists) have reinforced the synergy with cancer vaccines. Only limited results are available in humans and this combined approach has yet to be validated. Comprehensive monitoring of the regulation of CPI and costimulatory molecules after administration of immunomodulatory antibodies (anti-PD1/PD-L1, anti-CTLA-4, anti-OX40, etc.) and cancer vaccines should help to guide the selection of the best combination and timing of this therapy.
Collapse
|
15
|
Wu M, Li M, Yue Y, Xu W. DNA vaccine with discontinuous T-cell epitope insertions into HSP65 scaffold as a potential means to improve immunogenicity of multi-epitope Mycobacterium tuberculosis
vaccine. Microbiol Immunol 2016; 60:634-45. [DOI: 10.1111/1348-0421.12410] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/04/2016] [Accepted: 08/08/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Manli Wu
- Institute of Biology and Medical Sciences; Soochow University; Building 703, 199 Ren-ai Road Suzhou 215123 China
| | - Min Li
- Institute of Biology and Medical Sciences; Soochow University; Building 703, 199 Ren-ai Road Suzhou 215123 China
| | - Yan Yue
- Institute of Biology and Medical Sciences; Soochow University; Building 703, 199 Ren-ai Road Suzhou 215123 China
| | - Wei Xu
- Institute of Biology and Medical Sciences; Soochow University; Building 703, 199 Ren-ai Road Suzhou 215123 China
| |
Collapse
|
16
|
Haut LH, Gill AL, Kurupati RK, Bian A, Li Y, Giles-Davis W, Xiang Z, Zhou XY, Ertl HCJ. A Partial E3 Deletion in Replication-Defective Adenoviral Vectors Allows for Stable Expression of Potentially Toxic Transgene Products. Hum Gene Ther Methods 2016; 27:187-196. [PMID: 27604324 DOI: 10.1089/hgtb.2016.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Adenovirus (Ad) is used extensively for construction of viral vectors, most commonly with deletion in its E1 and/or E3 genomic regions. Previously, our attempts to insert envelope proteins (Env) of HIV-1 into such vectors based on chimpanzee-derived Ad (AdC) viruses were thwarted. Here, we describe that genetic instability of an E1- and E3-deleted AdC vector of serotype C6 expressing Env of HIV-1 can be overcome by reinsertion of E3 sequences with anti-apoptotic activities. This partial E3 deletion presumably delays premature death of HEK-293 packaging cell lines due to Env-induced cell apoptosis. The same partial E3 deletion also allows for the generation of stable glycoprotein 140 (gp140)- and gp160-expressing Ad vectors based on AdC7, a distinct AdC serotype. Env-expressing AdC vectors containing the partial E3 deletion are genetically stable upon serial cell culture passaging, produce yields comparable to those of other AdC vectors, and induce transgene product-specific antibody responses in mice. A partial E3 deletion thereby allows expansion of the repertoire of transgenes that can be expressed by Ad vectors.
Collapse
Affiliation(s)
| | - Amanda L Gill
- 1 The Wistar Institute , Philadelphia, Pennsylvania
- 2 Current address: Clinical Molecular Regulation Section/Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland
| | | | - Ang Bian
- 1 The Wistar Institute , Philadelphia, Pennsylvania
| | - Yan Li
- 1 The Wistar Institute , Philadelphia, Pennsylvania
| | | | | | | | | |
Collapse
|
17
|
Zhang Y, Ertl HC. Depletion of FAP+ cells reduces immunosuppressive cells and improves metabolism and functions CD8+T cells within tumors. Oncotarget 2016; 7:23282-99. [PMID: 26943036 PMCID: PMC5029626 DOI: 10.18632/oncotarget.7818] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/14/2016] [Indexed: 11/25/2022] Open
Abstract
The tumor stroma, which is essential to support growth and metastasis of malignant cells, provides targets for active immunotherapy of cancer. Previous studies have shown that depleting fibroblast activation protein (FAP)-expressing stromal cells reduces tumor progression and concomitantly increases tumor antigen (TA)-specific T cell responses. However the underlying pathways remain ill defined. Here we identify that immunosuppressive cells (ISCs) from tumor-bearing mice impose metabolic stress on CD8+T cells, which is associated with increased expression of the co-inhibitor PD-1. In two mouse melanoma models, depleting FAP+ stroma cells from the tumor microenvironment (TME) upon vaccination with an adenoviral-vector reduces frequencies and functions of ISCs. This is associated with changes in the cytokine/chemokine milieu in the TME and decreased activity of STAT6 signaling within ISCs. Decreases in ISCs upon FAP+stromal cell depletion is associated with reduced metabolic stress of vaccine-induced tumor infiltrating CD8+T cells and their delayed progression towards functional exhaustion, resulting in prolonged survival of tumor-bearing mice.
Collapse
Affiliation(s)
- Ying Zhang
- Gene Therapy and Vaccines Program, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
18
|
de Aquino MTP, Malhotra A, Mishra MK, Shanker A. Challenges and future perspectives of T cell immunotherapy in cancer. Immunol Lett 2015; 166:117-33. [PMID: 26096822 PMCID: PMC4499494 DOI: 10.1016/j.imlet.2015.05.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 05/10/2015] [Accepted: 05/27/2015] [Indexed: 12/15/2022]
Abstract
Since the formulation of the tumour immunosurveillance theory, considerable focus has been on enhancing the effectiveness of host antitumour immunity, particularly with respect to T cells. A cancer evades or alters the host immune response by various ways to ensure its development and survival. These include modifications of the immune cell metabolism and T cell signalling. An inhibitory cytokine milieu in the tumour microenvironment also leads to immune suppression and tumour progression within a host. This review traces the development in the field and attempts to summarize the hurdles that the approach of adoptive T cell immunotherapy against cancer faces, and discusses the conditions that must be improved to allow effective eradication of cancer.
Collapse
Affiliation(s)
- Maria Teresa P de Aquino
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Anshu Malhotra
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Manoj K Mishra
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; Tumor-Host Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
19
|
Baksh K, Weber J. Immune checkpoint protein inhibition for cancer: preclinical justification for CTLA-4 and PD-1 blockade and new combinations. Semin Oncol 2015; 42:363-77. [PMID: 25965355 DOI: 10.1053/j.seminoncol.2015.02.015] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Over the last two decades, our understanding of the molecular basis of immunity has revealed the complexity of regulatory pathways involved in immune responses to cancer. A significant body of data support the critical importance of immune checkpoints in the control of the adaptive immune response to malignancy, and suggest that inhibitors of those checkpoints might have significant utility in treating cancer. This has been borne out by the recent US Food and Drug Administration (FDA) approvals of two different antibodies, one against cytotoxic T-lymphocyte antigen-4 (CTLA-4) and one against programmed death-1 (PD-1). Here, we provide a comprehensive review of the literature regarding the preclinical justification for the use of CTLA-4 and PD-1 blockade as monotherapy, and as combination therapy in the treatment of cancer. The animal data strongly supported the use of these drugs in patients, and in many cases suggested strategies that directly led to successful registration trials. In contrast, many of the toxicities, and some of the unusual response patterns seen in patients with these drugs, were not predicted by the preclinical work that we cite, highlighting the importance of early-phase trials with patients to inform future drug development. In addition, we review herein the preclinical data surrounding emerging immune checkpoint proteins, including BTLA, VISTA, CD160, LAG3, TIM3, and CD244 as potential targets for inhibition. The current comprehensive review of the literature regarding CTLA-4 and PD-1, as well as a number of novel checkpoint proteins demonstrates a strong preclinical basis for the use of these antibodies singly and in combination to overcome checkpoint inhibition in the treatment of cancer. We also suggest that the use of these antibodies may augment the efficacy of other activating immune antibodies, cytokines, radiation, and adoptive cell therapy in human cancer.
Collapse
Affiliation(s)
- Kathryn Baksh
- Donald A. Adam Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL; University of South Florida School of Medicine, Tampa, FL.
| | - Jeffrey Weber
- Donald A. Adam Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|