113 results on '"Douglas G. McNeel"'
Search Results
2. GM-CSF elicits antibodies to tumor-associated proteins when used as a prostate cancer vaccine adjuvant
- Author
-
Hemanth K. Potluri, Tun L. Ng, Michael A. Newton, and Douglas G. McNeel
- Subjects
Male ,Vaccines ,Cancer Research ,Immunology ,Granulocyte-Macrophage Colony-Stimulating Factor ,Prostatic Neoplasms ,Cancer Vaccines ,Antibodies ,Neoplasm Proteins ,Adjuvants, Immunologic ,Oncology ,Humans ,Immunology and Allergy ,Adjuvants, Vaccine - Abstract
Antibody responses to off-target cancer-associated proteins have been detected following immunotherapies for cancer, suggesting these may be the result of antigen spread. We have previously reported that serum antibodies to prostate cancer-associated proteins were detectable using a high-throughput peptide array. We hypothesized that the breadth of antibody responses elicited by a vaccine could serve as a measure of the magnitude of its induced antigen spread. Consequently, sera from patients with prostate cancer, treated prior to or after vaccination in one of four separate clinical trials, were evaluated for antibody responses to an array of 177,604 peptides derived from over 1600 prostate cancer-associated gene products. Antibody responses to the same group of 5680 peptides previously reported were identified following vaccinations in which patients were administered GM-CSF as an adjuvant, but not with vaccine in the absence of GM-CSF. Hence, antibody responses to off-target proteins following vaccination may not necessarily serve as evidence of antigen spread and must be interpreted with particular caution following vaccine strategies that use GM-CSF, as GM-CSF appears to have direct effects on the production of antibodies. The evaluation of T cell responses to non-target antigens is likely a preferred approach for detection of immune-mediated antigen spread.
- Published
- 2022
3. Optimizing Flow Cytometric Analysis of Immune Cells in Samples Requiring Cryopreservation from Tumor-Bearing Mice
- Author
-
Jamey P. Weichert, Manasi Mohan, Christopher D. Zahm, Dagna Sheerar, Kathryn Fox, Zachary S. Morris, Douglas G. McNeel, Jen Birstler, Lauren Nettenstrom, Peter M. Carlson, Anna Hoefges, KyungMann Kim, Paul M. Sondel, Matthew Rodriguez, Ravi Patel, and Reinier Hernandez
- Subjects
CD4-Positive T-Lymphocytes ,Myeloid ,Immunology ,Melanoma, Experimental ,CD8-Positive T-Lymphocytes ,Biology ,Cryopreservation ,Immunophenotyping ,Flow cytometry ,Mice ,Immune system ,Cell Line, Tumor ,Novel Immunological Methods ,Tumor Microenvironment ,medicine ,Animals ,Immunology and Allergy ,Myeloid Cells ,Pandemics ,Fixation (histology) ,medicine.diagnostic_test ,Melanoma ,Flow Cytometry ,medicine.disease ,Staining ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Leukocytes, Mononuclear ,Cancer research ,Natural Killer T-Cells ,Signal Transduction - Abstract
Most shared resource flow cytometry facilities do not permit analysis of radioactive samples. We are investigating low-dose molecular targeted radionuclide therapy (MTRT) as an immunomodulator in combination with in situ tumor vaccines and need to analyze radioactive samples from MTRT-treated mice using flow cytometry. Further, the sudden shutdown of core facilities in response to the COVID-19 pandemic has created an unprecedented work stoppage. In these and other research settings, a robust and reliable means of cryopreservation of immune samples is required. We evaluated different fixation and cryopreservation protocols of disaggregated tumor cells with the aim of identifying a protocol for subsequent flow cytometry of the thawed sample, which most accurately reflects the flow cytometric analysis of the tumor immune microenvironment of a freshly disaggregated and analyzed sample. Cohorts of C57BL/6 mice bearing B78 melanoma tumors were evaluated using dual lymphoid and myeloid immunophenotyping panels involving fixation and cryopreservation at three distinct points during the workflow. Results demonstrate that freezing samples after all staining and fixation are completed most accurately matches the results from noncryopreserved equivalent samples. We observed that cryopreservation of living, unfixed cells introduces a nonuniform alteration to PD1 expression. We confirm the utility of our cryopreservation protocol by comparing tumors treated with in situ tumor vaccines, analyzing both fresh and cryopreserved tumor samples with similar results. Last, we use this cryopreservation protocol with radioactive specimens to demonstrate potentially beneficial effector cell changes to the tumor immune microenvironment following administration of a novel MTRT in a dose- and time-dependent manner.
- Published
- 2021
4. Role of B cells as antigen presenting cells
- Author
-
Ichwaku Rastogi, Donghwan Jeon, Jena E. Moseman, Anusha Muralidhar, Hemanth K. Potluri, and Douglas G. McNeel
- Subjects
Antigen Presentation ,B-Lymphocytes ,Immunology ,Immunology and Allergy ,Antigen-Presenting Cells ,Receptors, Antigen, B-Cell ,CD8-Positive T-Lymphocytes - Abstract
B cells have been long studied for their role and function in the humoral immune system. Apart from generating antibodies and an antibody-mediated memory response against pathogens, B cells are also capable of generating cell-mediated immunity. It has been demonstrated by several groups that B cells can activate antigen-specific CD4 and CD8 T cells, and can have regulatory and cytotoxic effects. The function of B cells as professional antigen presenting cells (APCs) to activate T cells has been largely understudied. This, however, requires attention as several recent reports have demonstrated the importance of B cells within the tumor microenvironment, and B cells are increasingly being evaluated as cellular therapies. Antigen presentation through B cells can be through antigen-specific (B cell receptor (BCR) dependent) or antigen non-specific (BCR independent) mechanisms and can be modulated by a variety of intrinsic and external factors. This review will discuss the pathways and mechanisms by which B cells present antigens, and how B cells differ from other professional APCs.
- Published
- 2022
5. Toll-like receptor agonist combinations augment mouse T-cell anti-tumor immunity via IL-12- and interferon ß-mediated suppression of immune checkpoint receptor expression
- Author
-
Donghwan, Jeon and Douglas G, McNeel
- Subjects
Programmed Cell Death 1 Receptor ,Toll-Like Receptors ,Immunology ,CD8-Positive T-Lymphocytes ,Immune Checkpoint Proteins ,Ligands ,Interleukin-12 ,Toll-Like Receptor 1 ,Toll-Like Receptor 3 ,Mice ,Adjuvants, Immunologic ,Oncology ,Toll-Like Receptor 9 ,Animals ,Immunology and Allergy ,Interferons - Abstract
We previously found that activated CD8
- Published
- 2022
6. 594 Combination of antigen-specific vaccination and targeted radionuclide therapy improves anti-tumor efficacy in a murine prostate model
- Author
-
Hemanth Potluri, Reinier Hernandez, Jamey P. Weichert, Carolina A. Ferreira, Douglas G. McNeel, Joseph Grudzinski, and Christopher Massey
- Subjects
Pharmacology ,Antitumor activity ,Cancer Research ,business.industry ,Immunology ,Targeted radionuclide therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Vaccination ,medicine.anatomical_structure ,Oncology ,Antigen specific ,Prostate ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 - Abstract
BackgroundWhile checkpoint blockade has been unsuccessful in prostate cancer trials, the approval of Sipuleucel-T demonstrates the value of antigen-specific vaccination approaches for this disease. We have studied a DNA vaccine specific for the ligand-binding domain of the androgen receptor (pTVG-AR) as a more scalable vaccination approach, though its efficacy is likely limited by the immunosuppressive prostate microenvironment. External beam radiotherapy has been shown to sensitize poorly responsive tumors to immunotherapy, but is infeasible for patients with widely metastatic disease. Our group has developed a compound called NM600 that can deliver radiation to all cancer sites simultaneously, similar to other targeted radionuclide therapy (TRT) approaches. In this study, we used TRT in combination with pTVG-AR to improve anti-tumor efficacy in a murine prostate cancer model.Methods6-week old male C57BL/6 mice were implanted subcutaneously with TRAMP-C1 cells. pTVG-AR or the empty vector were administered weekly from the day after tumor implantation. An intravenous injection was administered of 50 (”low-dose”) or 250 μCi (”high dose”) of 90Y-NM600, estimated to deliver a dose of 3.1 Gy or 15.5 Gy to 300 mm3 tumors, respectively. In one study, this TRT treatment was repeated once after three weeks. Groups of mice (n=5) were euthanized at several time points for flow cytometry analysis of the tumors. Separate cohorts (n=7) were followed for survival.ResultsLow-dose TRT administered once in combination with pTVG-AR (median survival 91 days) significantly improved survival more than low-dose TRT alone (median survival 59 days; p=.049) or pTVG-AR alone (median survival 59 days; p=0.01). Low-dose TRT plus pTVG-AR was also superior to high-dose TRT plus pTVG-AR (median survival 67 days; p=0.05). We next examined the effect of giving high-dose TRT twice in combination pTVG-AR. We found that the combination of fractionated TRT and pTVG-AR greatly slowed tumor growth unlike fractionated TRT alone (p=0.03). High-dose TRT + pTVG-AR caused a two-fold increase in CD86 expression on dendritic cells (p=0.0009) on Day 3 and a 10% increase in effector memory CD8+ T cells (p=0.002) on Day 1 compared to TRT alone. This combination also resulted in T cells with 3-fold lower PD-1 expression (p=4e-7) and 2-fold lower TIGIT expression (p=0.01).ConclusionsThese data suggest that the combination of antigen-specific vaccination and TRT can be an effective treatment for cancers that are refractory to immunotherapy. This combination may act through increasing co-stimulation by dendritic cells, leading to a more active cytolytic CD8+ T cell population.
- Published
- 2021
7. 350 Phase 2 trial of a DNA vaccine with pembrolizumab in patients with metastatic, castration-resistant prostate cancer (mCRPC)
- Author
-
Mary Jane Brennan, Laura E. Johnson, Christos Kyriakopoulos, Hamid Emamekhoo, Glenn Liu, Joshua M. Lang, Ellen Wargowski, Douglas G. McNeel, and Jens C. Eickhoff
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Immunology ,Pembrolizumab ,law.invention ,Prostate cancer ,Randomized controlled trial ,law ,Internal medicine ,Immunology and Allergy ,Medicine ,Adverse effect ,RC254-282 ,Pharmacology ,Hepatitis ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Rash ,Clinical trial ,Prostatic acid phosphatase ,Molecular Medicine ,medicine.symptom ,business - Abstract
BackgroundWe previously reported a pilot clinical trial using a DNA vaccine encoding prostatic acid phosphatase (pTVG-HP), given over 12 weeks either concurrently or in sequence with pembrolizumab, in patients with mCRPC. We report here the final analysis of this trial following two additional treatment arms in which patients with mCRPC were treated beyond 12 weeks until progression.MethodsPatients with mCRPC were treated with pTVG-HP and pembrolizumab every 3 weeks (Arm 3, n=20), or pTVG-HP every 2 weeks and pembrolizumab every 4 weeks (Arm 4, n=20). The primary objectives were safety, 6-month PFS, median time to radiographic progression, and objective response rates. Secondary objectives included immunological evaluations.ResultsTreatment was without unexpected toxicity, and only 1 grade 4 event (hyperglycemia) was observed. Immune related adverse events (irAE) > grade 1 included adrenal insufficiency, hepatitis, colitis, thyroid dysfunction, pancreatitis, pneumonitis, and rash, occurring in 42% of patients overall. 10/25 patients with measurable disease experienced any decrease in tumor volume from baseline, with 1 confirmed PR and no CR. 23/66 (35%) experienced any PSA decline from baseline. Overall median TTP was 5.4 months (95% CI; 5.3–8.1 months); median TTP for Arm 3 was 5.3 months compared to 8.0 months for Arm 4. Overall, 41.7% of patients had no radiographic progression at 6 months (29.9% Arm 3, 57.9% Arm 4). Median overall survival was 22.9 months. IFNγ and/or granzyme B immune response to PAP was detected in 2/20 patients in Arm 3 and 6/20 patients in Arm 4. Cytokines associated with immune activation and CD8+ T cell recruitment were augmented in the plasma of patients at weeks 6 and 12. Increased IFNγ in the sera at week 6 trended with prolonged TTP (p=0.010) and overall survival (p=0.025). The development of irAE was associated with a prolonged TTP (HR=0.25, p=0.003).ConclusionsPD-1 pathway inhibitors have demonstrated little clinical activity to date as monotherapies for mCRPC. Our findings demonstrate that combining PD-1 blockade with tumor-targeted T-cell activation using pTVG-HP is safe, can augment tumor-specific T cells, and result in objective changes with longer time to progression than what has been observed in previous trials. The association of progression or survival with increased IFNγ, irAE, and vaccine schedule suggests T cell activation by vaccination is critical to the mechanism of action of this combination. This study suggests this approach should be further evaluated in randomized clinical trials for patients with advanced mCRPC.AcknowledgementsFunding for this trial was from a 2014 Movember Prostate Cancer Foundation Challenge Award and Madison Vaccines, Inc.Trial RegistrationNCT02499835Ethics ApprovalThis trial was reviewed and approved by the University of Wisconsin Human Subjects’ Review Committee (IRB), protocol 2015–0453. All participants provided IRB-approved written informed consent before taking part.
- Published
- 2021
8. Antitumor efficacy of 90Y-NM600 targeted radionuclide therapy and PD-1 blockade is limited by regulatory T cells in murine prostate tumors
- Author
-
Hemanth K Potluri, Carolina A Ferreira, Joseph Grudzinski, Christopher Massey, Eduardo Aluicio-Sarduy, Jonathan W Engle, Ohyun Kwon, Ian R Marsh, Bryan P Bednarz, Reinier Hernandez, Jamey P Weichert, and Douglas G McNeel
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Immunology ,Molecular Medicine ,Immunology and Allergy - Abstract
BackgroundSystemic radiation treatments that preferentially irradiate cancer cells over normal tissue, known as targeted radionuclide therapy (TRT), have shown significant potential for treating metastatic prostate cancer. Preclinical studies have demonstrated the ability of external beam radiation therapy (EBRT) to sensitize tumors to T cell checkpoint blockade. Combining TRT approaches with immunotherapy may be more feasible than combining with EBRT to treat widely metastatic disease, however the effects of TRT on the prostate tumor microenvironment alone and in combinfation with checkpoint blockade have not yet been studied.MethodsC57BL/6 mice-bearing TRAMP-C1 tumors and FVB/NJ mice-bearing Myc-CaP tumors were treated with a single intravenous administration of either low-dose or high-dose 90Y-NM600 TRT, and with or without anti-PD-1 therapy. Groups of mice were followed for tumor growth while others were used for tissue collection and immunophenotyping of the tumors via flow cytometry.Results90Y-NM600 TRT was safe at doses that elicited a moderate antitumor response. TRT had multiple effects on the tumor microenvironment including increasing CD8 +T cell infiltration, increasing checkpoint molecule expression on CD8 +T cells, and increasing PD-L1 expression on myeloid cells. However, PD-1 blockade with TRT treatment did not improve antitumor efficacy. Tregs remained functional up to 1 week following TRT, but CD8 +T cells were not, and the suppressive function of Tregs increased when anti-PD-1 was present in in vitro studies. The combination of anti-PD-1 and TRT was only effective in vivo when Tregs were depleted.ConclusionsOur data suggest that the combination of 90Y-NM600 TRT and PD-1 blockade therapy is ineffective in these prostate cancer models due to the activating effect of anti-PD-1 on Tregs. This finding underscores the importance of thorough understanding of the effects of TRT and immunotherapy combinations on the tumor immune microenvironment prior to clinical investigation.
- Published
- 2022
9. PD-1 and LAG-3 blockade improve anti-tumor vaccine efficacy
- Author
-
Douglas G. McNeel, Jena E Moseman, Christopher D. Zahm, and Lauren E. Delmastro
- Subjects
0301 basic medicine ,Male ,Immunology ,Programmed Cell Death 1 Receptor ,Pharmacology ,CD8-Positive T-Lymphocytes ,Cancer Vaccines ,03 medical and health sciences ,Mice ,0302 clinical medicine ,LAG-3 ,Antigens, CD ,Neoplasms ,PD-1 ,Vaccines, DNA ,Immunology and Allergy ,Medicine ,Animals ,Receptor ,RC254-282 ,Original Research ,Antitumor activity ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC581-607 ,Vaccine efficacy ,Lymphocyte Activation Gene 3 Protein ,Blockade ,APC ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,tumor vaccine ,biological phenomena, cell phenomena, and immunity ,Immunologic diseases. Allergy ,business ,Research Article - Abstract
Concurrent blockade of different checkpoint receptors, notably PD-1 and CTLA-4, elicits greater anti-tumor activity for some tumor types, and the combination of different checkpoint receptor inhibitors is an active area of clinical research. We have previously demonstrated that anti-tumor vaccination, by activating CD8 + T cells, increases the expression of PD-1, CTLA-4, LAG-3 and other inhibitory receptors, and the anti-tumor efficacy of vaccination can be increased with checkpoint blockade. In the current study, we sought to determine whether anti-tumor vaccination might be further improved with combined checkpoint blockade. Using an OVA-expressing mouse tumor model, we found that CD8 + T cells activated in the presence of professional antigen presenting cells (APC) expressed multiple checkpoint receptors; however, T cells activated without APCs expressed LAG-3 alone, suggesting that LAG-3 might be a preferred target in combination with vaccination. Using three different murine tumor models, and peptide or DNA vaccines targeting three tumor antigens, we assessed the effects of vaccines with blockade of PD-1 and/or LAG-3 on tumor growth. We report that, in each model, the anti-tumor efficacy of vaccination was increased with PD-1 and/or LAG-3 blockade. However, combined PD-1 and LAG-3 blockade elicited the greatest anti-tumor effect when combined with vaccination in a MycCaP prostate cancer model in which PD-1 blockade alone with vaccination targeting a “self” tumor antigen had less efficacy. These results suggest anti-tumor vaccination might best be combined with concurrent blockade of both PD-1 and LAG-3, and potentially other checkpoint receptors whose expression is increased on CD8 + T cells following vaccine-mediated activation.
- Published
- 2021
10. B cells licensed by DCs are primary antigen presenting cells for DNA plasmid upon passive uptake
- Author
-
Ichwaku Rastogi and Douglas G McNeel
- Subjects
Immunology ,Immunology and Allergy - Abstract
Dendritic cells are considered the primary cells involved in professional antigen presentation. However, our group and others have shown that B cells are similarly capable of presenting antigens leading to activation and proliferation of antigen-specific T cells. In this study, we aimed to understand the mechanism of presentation of antigen, encoded by plasmid DNA when delivered to APCs via passive uptake. We used plasmid DNA encoding ovalbumin, splenic B cells from Bl6 mice, DCs from Flt3L treated Bl6 mice, and CD8 T cells from OT1 mice that are restricted for the SIINFEKL-specific T-cell receptor. We have previously reported that only B cells are capable of transcribing the encoded antigen, whereas DCs and macrophages degrade the plasmid DNA upon passive uptake. In our current study we show that B cells also translate the encoded antigen, but can present the antigen to CD8 T cells only in the presence of DCs. We utilized a trans-well assay to show that physical contact between B cells and DCs is required for successful antigen presentation and that live DCs are essential, as replacing them with DC lysate or DC supernatant or both was not adequate. We also show that the presence of DCs promotes B cell survival, mostly through CD40-CD40L signaling by using CD40 KO mice and that B cells are primary antigen presenting cells in this co-culture by using MHC I KO mice. We also show that exposure of B cells to CD40 or CD40L, which is one of the identified interactions between B cells and DCs, was not sufficient for B cells to present antigen in the absence of DCs. In summary, our data suggests that B cells and DCs communicate through a currently unknown cell-cell interaction that licenses B cells for antigen presentation after passive uptake of plasmid DNA.
- Published
- 2022
11. Phase 2 trial of T-cell activation using MVI-816 and pembrolizumab in patients with metastatic, castration-resistant prostate cancer (mCRPC)
- Author
-
Douglas G McNeel, Jens C Eickhoff, Ellen Wargowski, Laura E Johnson, Christos E Kyriakopoulos, Hamid Emamekhoo, Joshua M Lang, Mary Jane Brennan, and Glenn Liu
- Subjects
Male ,Pharmacology ,Prostatic Neoplasms, Castration-Resistant ,Cancer Research ,Oncology ,Immunology ,Vaccines, DNA ,Humans ,Molecular Medicine ,Immunology and Allergy ,Antibodies, Monoclonal, Humanized ,Progression-Free Survival - Abstract
BackgroundWe previously reported a trial using a DNA vaccine encoding prostatic acid phosphatase (MVI-816, pTVG-HP), given over 12 weeks concurrently or sequentially with pembrolizumab, in patients with mCRPC. We report the final analysis of this trial following two additional treatment arms in which patients with mCRPC continued concurrent treatment until progression.Materials and methodsPatients with mCRPC were treated with MVI-816 and pembrolizumab every 3 weeks (arm 3, n=20) or MVI-816 every 2 weeks and pembrolizumab every 4 weeks (arm 4, n=20). The primary objectives were safety, 6-month progression-free survival (PFS), median time to radiographic progression, and objective response rates. Secondary objectives included immunological evaluations.ResultsIn 25 patients with measurable disease, there were no complete response and one confirmed partial response in a patient who subsequently found to have an MSIhitumor. 4/40 patients (10%) had a prostate-specific antigen decline >50%. The estimated overall radiographic PFS rate at 6 months was 47.2% (44.4% arm 3, 61.5% arm 4). Accounting for all off-study events, overall median time on treatment was 5.6 months (95% CI: 5.4 to 10.8 months), 5.6 months for arm 3 and 8.1 months for arm 4 (p=0.64). Thirty-two per cent of patients remained on trial beyond 6 months without progression. Median overall survival was 22.9 (95% CI: 16.2 to 25.6) months. One grade 4 event (hyperglycemia) was observed. Immune-related adverse events (irAEs) >grade 1 were observed in 42% of patients overall. Interferon-γ and/or granzyme B immune response to prostatic acid phosphatase was detected in 2/20 patients in arm 3 and 6/20 patients in arm 4. Plasma cytokines associated with immune activation and CD8+ T-cell recruitment were augmented at weeks 6 and 12. The development of irAE was significantly associated with a prolonged time on treatment (HR=0.42, p=0.003). Baseline DNA homologous recombination repair mutations were not associated with longer time to progression.ConclusionsFindings here demonstrate that combining programmed cell death 1 blockade with MVI-816 is safe, can augment tumor-specific T cells, and can result in a favorable 6-month disease control rate. Correlative studies suggest T-cell activation by vaccination is critical to the mechanism of action of this combination. Future randomized clinical trials are needed to validate these findings.Trial registration numberNCT02499835.
- Published
- 2022
12. Immune system and intestinal microbiota determine efficacy of androgen deprivation therapy against prostate cancer
- Author
-
Safae Terrisse, Anne-Gaelle Goubet, Kousuke Ueda, Andrew Maltez Thomas, Valentin Quiniou, Cassandra Thelemaque, Garett Dunsmore, Emmanuel Clave, Melissa Gamat-Huber, Satoru Yonekura, Gladys Ferrere, Conrad Rauber, Hang Phuong Pham, Jean-Eudes Fahrner, Eugenie Pizzato, Pierre Ly, Marine Fidelle, Marine Mazzenga, Carolina Alves Costa Silva, Federica Armanini, Federica Pinto, Francesco Asnicar, Romain Daillère, Lisa Derosa, Corentin Richard, Pierre Blanchard, Bertrand Routy, Stéphane Culine, Paule Opolon, Aymeric Silvin, Florent Ginhoux, Antoine Toubert, Nicola Segata, Douglas G McNeel, Karim Fizazi, Guido Kroemer, and Laurence Zitvogel
- Subjects
Male ,Pharmacology ,Cancer Research ,Immunology ,Prostatic Neoplasms ,Androgen Antagonists ,adaptive immunity ,immunomodulation ,Castration-Resistant ,Gastrointestinal Microbiome ,Prostatic Neoplasms, Castration-Resistant ,Mice ,prostatic neoplasms ,translational medical research ,Androgens ,Animals ,Humans ,Immune System ,Oncology ,Molecular Medicine ,Immunology and Allergy - Abstract
BackgroundProstate cancer (PC) responds to androgen deprivation therapy (ADT) usually in a transient fashion, progressing from hormone-sensitive PC (HSPC) to castration-resistant PC (CRPC). We investigated a mouse model of PC as well as specimens from PC patients to unravel an unsuspected contribution of thymus-derived T lymphocytes and the intestinal microbiota in the efficacy of ADT.MethodsPreclinical experiments were performed in PC-bearing mice, immunocompetent or immunodeficient. In parallel, we prospectively included 65 HSPC and CRPC patients (Oncobiotic trial) to analyze their feces and blood specimens.ResultsIn PC-bearing mice, ADT increased thymic cellularity and output. PC implanted in T lymphocyte-depleted or athymic mice responded less efficiently to ADT than in immunocompetent mice. Moreover, depletion of the intestinal microbiota by oral antibiotics reduced the efficacy of ADT. PC reduced the relative abundance of Akkermansia muciniphila in the gut, and this effect was reversed by ADT. Moreover, cohousing of PC-bearing mice with tumor-free mice or oral gavage with Akkermansia improved the efficacy of ADT. This appears to be applicable to PC patients because long-term ADT resulted in an increase of thymic output, as demonstrated by an increase in circulating recent thymic emigrant cells (sjTRECs). Moreover, as compared with HSPC controls, CRPC patients demonstrated a shift in their intestinal microbiota that significantly correlated with sjTRECs. While feces from healthy volunteers restored ADT efficacy, feces from PC patients failed to do so.ConclusionsThese findings suggest the potential clinical utility of reversing intestinal dysbiosis and repairing acquired immune defects in PC patients.
- Published
- 2022
13. Infectious Tolerance as Seen With 2020 Vision: The Role of IL-35 and Extracellular Vesicles
- Author
-
Brian Olson, Jeremy A. Sullivan, Douglas G. McNeel, William J. Burlingham, and David P. Al-Adra
- Subjects
lcsh:Immunologic diseases. Allergy ,medicine.medical_treatment ,Lymphocyte ,Immunology ,Review ,exosomes ,Biology ,History, 21st Century ,Extracellular vesicles ,Extracellular Vesicles ,Allergy and Immunology ,Immune Tolerance ,medicine ,Animals ,Humans ,Immunology and Allergy ,Mechanism (biology) ,Interleukins ,Peripheral tolerance ,Immunotherapy ,Extracellular vesicle ,History, 20th Century ,infectious tolerance ,Microvesicles ,Cytokine ,medicine.anatomical_structure ,IL-35 ,extracellular vesicle ,immunotherapy ,lcsh:RC581-607 - Abstract
Originally identified as lymphocyte regulation of fellow lymphocytes, our understanding of infectious tolerance has undergone significant evolutions in understanding since being proposed in the early 1970s by Gershon and Kondo and expanded upon by Herman Waldman two decades later. The evolution of our understanding of infectious tolerance has coincided with significant cellular and humoral discoveries. The early studies leading to the isolation and identification of Regulatory T cells (Tregs) and cytokines including TGFβ and IL-10 in the control of peripheral tolerance was a paradigm shift in our understanding of infectious tolerance. More recently, another potential, paradigm shift in our understanding of the “infectious” aspect of infectious tolerance was proposed, identifying extracellular vesicles (EVs) as a mechanism for propagating infectious tolerance. In this review, we will outline the history of infectious tolerance, focusing on a potential EV mechanism for infectious tolerance and a novel, EV-associated form for the cytokine IL-35, ideally suited to the task of propagating tolerance by “infecting” other lymphocytes.
- Published
- 2020
- Full Text
- View/download PDF
14. Insights from immuno-oncology: the Society for Immunotherapy of Cancer Statement on access to IL-6-targeting therapies for COVID-19
- Author
-
Charles G. Drake, Walter J. Urba, David Kaufman, Leisha A. Emens, Jeffrey S. Weber, Alessandra Cesano, Douglas G. McNeel, Ana C. Anderson, Marcela V. Maus, Mario Sznol, Jon M Wiggington, Lisa H. Butterfield, Patrick Hwu, David Parkinson, Ernest C. Borden, James L. Gulley, Michael J. Mastrangelo, Michael B. Atkins, Kim Margolin, Julie R. Brahmer, Thomas F. Gajewski, Robert O. Dillman, Pedro Romero, Daniel S. Chen, Francesco M. Marincola, George J. Weiner, Paolo A. Ascierto, Tanja D. de Gruijl, Michael T. Lotze, Paul M. Sondel, F. Stephen Hodi, Bernard A. Fox, Stefani Spranger, and Howard L. Kaufman
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,viruses ,medicine.medical_treatment ,Immunology ,Betacoronavirus ,Coronavirus Infections/diagnostic imaging ,Coronavirus Infections/drug therapy ,Coronavirus Infections/immunology ,Coronavirus Infections/therapy ,Drug Approval ,Humans ,Immunologic Factors/therapeutic use ,Immunotherapy ,Inflammation/blood ,Inflammation/therapy ,Interleukin-6/agonists ,Neoplasms/therapy ,Pandemics ,Pneumonia, Viral/diagnostic imaging ,Pneumonia, Viral/drug therapy ,Pneumonia, Viral/immunology ,Pneumonia, Viral/therapy ,Receptors, Interleukin-6/agonists ,immunomodulation ,inflammation mediators ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Intensive care ,Pandemic ,medicine ,Immunology and Allergy ,Interleukin 6 ,RC254-282 ,Pneumonitis ,Pharmacology ,biology ,business.industry ,virus diseases ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Hypoxia (medical) ,medicine.disease ,030104 developmental biology ,Editorial ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Respiratory virus ,medicine.symptom ,business - Abstract
The hypoxia and profound inflammatory response associated with the pneumonitis observed with the severe acute respiratory virus coronavirus-2 (SARS-COV-2) virus responsible for the recent COVID-19 pandemic has overwhelmed intensive care facilities in the epicenters of infection including Wuhan
- Published
- 2020
15. TLR Stimulation during T-cell Activation Lowers PD-1 Expression on CD8+ T Cells
- Author
-
Viswa Teja Colluru, Irene M. Ong, Douglas G. McNeel, Christopher D. Zahm, and Sean J. McIlwain
- Subjects
0301 basic medicine ,Cancer Research ,Adoptive cell transfer ,Ovalbumin ,T cell ,Programmed Cell Death 1 Receptor ,Immunology ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Article ,Interleukin-12 Subunit p35 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Cytotoxic T cell ,Innate immune system ,Chemistry ,Toll-Like Receptors ,Imidazoles ,TLR9 ,Neoplasms, Experimental ,TLR7 ,Adoptive Transfer ,Peptide Fragments ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Aminoquinolines ,Cancer research ,Interleukin 12 ,Female ,CD8 - Abstract
Expression of T-cell checkpoint receptors can compromise antitumor immunity. Blockade of these receptors, notably PD-1 and LAG-3, which become expressed during T-cell activation with vaccination, can improve antitumor immunity. We evaluated whether T-cell checkpoint expression could be separated from T-cell activation in the context of innate immune stimulation with TLR agonists. We found that ligands for TLR1/2, TLR7, and TLR9 led to a decrease in expression of PD-1 on antigen-activated CD8+ T cells. These effects were mediated by IL12 released by professional antigen-presenting cells. In two separate tumor models, treatment with antitumor vaccines combined with TLR1/2 or TLR7 ligands induced antigen-specific CD8+ T cells with lower PD-1 expression and improved antitumor immunity. These findings highlight the role of innate immune activation during effector T-cell development and suggest that at least one mechanism by which specific TLR agonists can be strategically used as vaccine adjuvants is by modulating the expression of PD-1 during CD8+ T-cell activation. Cancer Immunol Res; 6(11); 1364–74. ©2018 AACR.
- Published
- 2018
16. Prime-boost vaccination targeting prostatic acid phosphatase (PAP) in patients with metastatic castration-resistant prostate cancer (mCRPC) using Sipuleucel-T and a DNA vaccine
- Author
-
Glenn Liu, Douglas G. McNeel, Ellen Wargowski, Jens C. Eickhoff, Lauren E. Delmastro, Mary Jane Staab, and Laura E. Johnson
- Subjects
Male ,Oncology ,DNA vaccine ,Cancer Research ,medicine.medical_specialty ,Acid Phosphatase ,Immunology ,Sipuleucel-T ,Prostatic acid phosphatase ,Cancer Vaccines ,lcsh:RC254-282 ,DNA vaccination ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Antigens, Neoplasm ,Prostate ,Immunity ,Internal medicine ,Vaccines, DNA ,medicine ,Humans ,Immunology and Allergy ,030212 general & internal medicine ,Immune monitoring ,Aged ,Aged, 80 and over ,Pharmacology ,Tissue Extracts ,business.industry ,Vaccination ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Clinical trial ,Prostatic Neoplasms, Castration-Resistant ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Molecular Medicine ,business ,Research Article ,medicine.drug - Abstract
Background Prostatic acid phosphatase (PAP) is a prostate tumor antigen, and the target of the only FDA-approved anti-tumor vaccine, sipuleucel-T. We have previously reported in two clinical trials that a DNA vaccine encoding PAP (pTVG-HP) could elicit PAP-specific, Th1-biased T cells in patients with PSA-recurrent prostate cancer. In the current pilot trial we sought to evaluate whether this vaccine could augment PAP-specific immunity when used as a booster to immunization with sipuleucel-T in patients with metastatic, castration-resistant prostate cancer (mCRPC). Methods Eigthteen patients with mCRPC were randomized to receive sipuleucel-T alone or followed by intradermal immunization with pTVG-HP DNA vaccine. Patients were followed for time to progression, and immune monitoring was conducted at defined intervals. Results Overall, patients were followed for a median of 24 months. 11/18 patients completed treatments as per protocol. No treatment-associated events > grade 2 were observed. Th1-biased PAP-specific T-cell responses were detected in 11/18 individuals, and were not statistically different between study arms. Higher titer antibody responses to PAP were detectable in patients who received pTVG-HP booster immunizations. Median time to progression was less than 6 months and not statistically different between study arms. The median overall survival for all patients was 28 months. Conclusions These findings suggest that prime-boost vaccination can augment and diversify the type of immunity elicited with anti-tumor vaccination in terms of T-cell and humoral immunity. Future studies will explore DNA as priming immunization rather than a booster immunization. Trial registration NCT01706458.
- Published
- 2018
17. Immunological considerations underlying heat shock protein-mediated cancer vaccine strategies
- Author
-
Miroslav Malkovsky, Matthew Kelly, Paul Fisch, and Douglas G. McNeel
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Active immunotherapy ,Biology ,Cancer Vaccines ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,Antigens, Neoplasm ,Neoplasms ,Heat shock protein ,Vaccines, DNA ,medicine ,Animals ,Humans ,Immunology and Allergy ,Heat-Shock Proteins ,Cancer ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunotherapy ,Cancer vaccine ,Intracellular - Abstract
The success of active immunotherapies in the prevention of many infectious diseases over the course of over 200 years has lead scientists to wonder if the same principles could be applied to cancer. Antigen-specific active immunotherapies for the treatment of cancer have been researched for over two decades, however, the overwhelming majority of these studies have failed to stimulate robust clinical responses. It is clear that current active immunotherapy research should incorporate methods to increase the immunostimulatory capacity of these therapies. To directly address this need, we propose the addition of the immunostimulatory heat shock proteins (HSPs) to active immunotherapeutic strategies to augment their efficacy. Heat shock proteins are a family of highly conserved intracellular chaperone proteins, and are the most abundant family proteins inside cells. This ubiquity, and their robust immunostimulatory capacity, points to their importance in regulation of intracellular processes and, therefore, indicators of loss of cellular integrity if found extracellularly. Thus, we emphasize the importance of taking into consideration the location of vaccine-derived HSP/tumor-antigen complexes when designing active immunotheraputic strategies.
- Published
- 2018
18. 767 Activation of CD8+ T cells in the presence of multiple TLR agonists affects the expression of T-cell checkpoint receptors via IL-12 and type-1 interferon
- Author
-
Donghwan Jeon and Douglas G. McNeel
- Subjects
Pharmacology ,Cancer Research ,Chemistry ,T cell ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Tlr agonists ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Interleukin 12 ,Molecular Medicine ,Immunology and Allergy ,Cytotoxic T cell ,Receptor ,Type 1 interferon ,RC254-282 - Abstract
BackgroundT-cell checkpoint receptors are expressed when T-cell are activated, and activation of these receptors can impair the function of T-cells and their anti-tumor efficacy.1 We previously found that T-cells activated with cognate antigen increase the expression of PD-1, while this can be attenuated by the presence of specific Toll-like receptor (TLR) agonists.2 3 This effect was mediated by IL-12 secretion from professional antigen presenting cells and resulted in CD8+ T cells with greater anti-tumor activity. In the current report, we sought to determine whether combination of TLR agonists can further affect the expression of T-cell checkpoint receptors and improve T-cell anti-tumor immunity.MethodsOT-1 CD8+ T cells were stimulated with peptide (SIINFEKL) and dendritic cells (DC) in the presence of two different TLR agonists. The cells were collected and evaluated for the expression of T-cell checkpoint receptors (PD-1, CTLA-4, CD160, CD244, LAG-3, TIM-3, TIGIT and VISTA) by flow cytometry, and for transcriptional changes by RNA-seq. Purified DC were stimulated with TLR combinations and evaluated for cytokine release by ELISA. The anti-tumor efficacy of vaccination using peptide and TLR agonist combinations was evaluated in EG7-OVA tumor-bearing mice.ResultsActivation of CD8+ T cells in the presence of specific TLR ligands resulted in decreases in expression of PD-1 and/or CD160. These changes in T-cell checkpoint receptor expression were modestly affected when TLR ligands were used in combination, and notably with combinations of TLR1/2, TLR3, and TLR9 agonists. Immunization of tumor-bearing mice, co-administered with combinations of these agonists, showed greater anti-tumor effects. However, while the effect of TLR1/2 and/or TLR9 was abrogated in IL12KO mice, TLR3 demonstrated anti-tumor activity when co-administered with peptide vaccine. RNA sequencing of TLR-conditioned CD8+ T-cells revealed IL-12 pathway activation, and IFNß pathway activation following TLR3 stimulation. Stimulation of DC with TLR3 agonist, alone or in combination with other TLR agonists, resulted in increased IL-12 and IFNß secretion. Co-incubation of OT-1 splenocytes with rIL12 and/or rIFNß during peptide activation led to reduced expression of PD-1, and this could be reversed with antibodies blocking IL12R or IFNAR-1.ConclusionsMultiple TLR agonists can modulate the expression of T-cell checkpoint receptors, notably PD-1, by upregulating the secretion of IL-12 and IFNß. These data provide the mechanistic rationale for choosing optimal combinations of TLR ligands to use as adjuvants to improve the efficacy of anti-tumor vaccines.ReferencesJin H-T, et al. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proceedings of the National Academy of Sciences 2010;107(33):14733–14738.Zahm CD, Colluru VT, McNeel DG. Vaccination with high-affinity epitopes impairs antitumor efficacy by increasing PD-1 expression on CD8+ T cells. Cancer Immunology Research 2017;5(8):630–641.Zahm CD, et al. TLR stimulation during T-cell activation lowers PD-1 expression on CD8+ T Cells. Cancer Immunology Research 2018;6(11):1364–1374.
- Published
- 2021
19. Prostate Cancer Cells Express More Androgen Receptor (AR) Following Androgen Deprivation, Improving Recognition by AR-Specific T Cells
- Author
-
Douglas G. McNeel, Charles G. Drake, Brian M. Olson, Jamey P. Weichert, Joseph Seliski, Thomas Sawicki, Melissa Gamat, Leigh Ellis, and Justin J. Jeffery
- Subjects
Male ,0301 basic medicine ,Cancer Research ,medicine.drug_class ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,T-Cell Antigen Receptor Specificity ,Article ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,In vivo ,Cell Line, Tumor ,Positron Emission Tomography Computed Tomography ,medicine ,Animals ,Humans ,Receptor ,business.industry ,Gene Expression Profiling ,Prostatic Neoplasms ,Cancer ,Immunotherapy ,medicine.disease ,Androgen ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Cancer research ,Immunization ,business - Abstract
Androgen deprivation is the primary therapy for recurrent prostate cancer, and agents targeting the androgen receptor (AR) pathway continue to be developed. Because androgen-deprivation therapy (ADT) has immmunostimulatory effects as well as direct antitumor effects, AR-targeted therapies have been combined with other anticancer therapies, including immunotherapies. Here, we sought to study whether an antigen-specific mechanism of resistance to ADT (overexpression of the AR) may result in enhanced AR-specific T-cell immune recognition, and whether this might be strategically combined with an antitumor vaccine targeting the AR. Androgen deprivation increased AR expression in human and murine prostate tumor cells in vitro and in vivo. The increased expression persisted over time. Increased AR expression was associated with recognition and cytolytic activity by AR-specific T cells. Furthermore, ADT combined with vaccination, specifically a DNA vaccine encoding the ligand-binding domain of the AR, led to improved antitumor responses as measured by tumor volumes and delays in the emergence of castrate-resistant prostate tumors in two murine prostate cancer models (Myc-CaP and prostate-specific PTEN-deficient mice). Together, these data suggest that ADT combined with AR-directed immunotherapy targets a major mechanism of resistance, overexpression of the AR. This combination may be more effective than ADT combined with other immunotherapeutic approaches. Cancer Immunol Res; 5(12); 1074–85. ©2017 AACR.
- Published
- 2017
20. DNA vaccines for prostate cancer
- Author
-
Douglas G. McNeel, Viswa Teja Colluru, and Christopher D. Zahm
- Subjects
Male ,0301 basic medicine ,medicine.medical_treatment ,Antigen-Presenting Cells ,Context (language use) ,Biology ,Cancer Vaccines ,Article ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Vaccines, DNA ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Treatment Failure ,Pharmacology ,Immunogenicity ,Prostatic Neoplasms ,Cancer ,Immunotherapy ,medicine.disease ,Immunity, Innate ,Vaccination ,Treatment Outcome ,030104 developmental biology ,Immunology ,030215 immunology - Abstract
DNA vaccines offer many advantages over other anti-tumor vaccine approaches due to their simplicity, ease of manufacturing, and safety. Results from several clinical trials in patients with cancer have demonstrated that DNA vaccines are safe and can elicit immune responses. However, to date few DNA vaccines have progressed beyond phase I clinical trial evaluation. Studies into the mechanism of action of DNA vaccines in terms of antigen-presenting cell types able to directly present or cross-present DNA-encoded antigens, and the activation of innate immune responses due to DNA itself, have suggested opportunities to increase the immunogenicity of these vaccines. In addition, studies into the mechanisms of tumor resistance to anti-tumor vaccination have suggested combination approaches that can increase the anti-tumor effect of DNA vaccines. This review focuses on these mechanisms of action and mechanisms of resistance using DNA vaccines, and how this information is being used to improve the anti-tumor effect of DNA vaccines. These approaches are then specifically discussed in the context of human prostate cancer, a disease for which DNA vaccines have been and continue to be explored as treatments.
- Published
- 2017
21. FLT PET/CT imaging of metastatic prostate cancer patients treated with pTVG-HP DNA vaccine and pembrolizumab
- Author
-
Robert Jeraj, Matthew Scarpelli, Glenn Liu, Christopher D. Zahm, Douglas G. McNeel, and Scott B. Perlman
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,medicine.medical_treatment ,Pembrolizumab ,Imaging ,Prostate cancer ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Positron Emission Tomography Computed Tomography ,Vaccines, DNA ,Immunology and Allergy ,Cell proliferation ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Clinical trial ,Prostatic Neoplasms, Castration-Resistant ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Molecular Medicine ,Biomarker (medicine) ,Lymph ,Thyroid function ,Research Article ,DNA vaccine ,medicine.medical_specialty ,Immunology ,Antibodies, Monoclonal, Humanized ,lcsh:RC254-282 ,03 medical and health sciences ,Response assessment ,Internal medicine ,medicine ,Humans ,Adverse effect ,Pharmacology ,FLT PET ,business.industry ,Immunotherapy ,medicine.disease ,Dideoxynucleosides ,030104 developmental biology ,Adverse events ,Bone marrow ,sense organs ,business - Abstract
Background Immunotherapy has demonstrated remarkable success in treating different cancers. Nonetheless, a large number of patients do not respond, many respond without immediate changes detectable with conventional imaging, and many have unusual immune-related adverse events that cannot be predicted in advance. In this exploratory study, we investigate how 3′-Deoxy-3’-18F-fluorothymidine (FLT) positron emission tomography (PET) measurements of tumor and immune cell proliferation might be utilized as biomarkers in immunotherapy. Methods Seventeen patients with metastatic castrate resistant prostate cancer were treated with combination pTVG-HP DNA vaccine and pembrolizumab. Patients underwent baseline and 12-week FLT PET/CT scans. FLT PET standardized uptake values (SUVs) were extracted from tumors, non-metastatic lymph nodes, spleen, bone marrow, pancreas, and thyroid to quantify cell proliferation in these tissues. Regional immune cell responses to pTVG-HP DNA vaccine were assessed by comparing FLT uptake changes in vaccine draining and non-draining lymph nodes. Cox proportional hazards regression was utilized to relate FLT uptake and other clinical markers (PSA and tumor size) to progression-free survival. Area under receiver operating characteristic (AUC) curves and concordance indices were used to assess the predictive capabilities of FLT uptake. Results Changes in FLT uptake in vaccine draining lymph nodes were significantly greater than changes in non-draining lymph nodes (P = 0.02), suggesting a regional immune response to vaccination. However, the changes in FLT uptake in lymph nodes were not significantly predictive of progression-free survival. Increases in tumor FLT uptake were significantly predictive of shorter progression-free survival (concordance index = 0.83, P
- Published
- 2018
22. Antibody profiling of patients with prostate cancer reveals differences in antibody signatures among disease stages
- Author
-
Peter S. Nelson, Tun Lee Ng, Michael A. Newton, Hemanth Potluri, Douglas G. McNeel, Jin Zhang, and Christopher G. Maher
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Immunology ,Antibodies ,Androgen deprivation therapy ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Antigen ,antigens ,Prostate ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,RC254-282 ,Neoplasm Staging ,Clinical/Translational Cancer Immunotherapy ,Pharmacology ,biology ,business.industry ,Autoantibody ,Prostatic Neoplasms ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,vaccination ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Prostatic acid phosphatase ,030220 oncology & carcinogenesis ,Disease Progression ,biology.protein ,Molecular Medicine ,Peptide microarray ,Antibody ,business ,neoplasm - Abstract
BackgroundPrevious studies of prostate cancer autoantibodies have largely focused on diagnostic applications. So far, there have been no reports attempting to more comprehensively profile the landscape of prostate cancer-associated antibodies. Specifically, it is unknown whether the quantity of antibodies or the types of proteins recognized change with disease progression.MethodsA peptide microarray spanning the amino acid sequences of the gene products of 1611 prostate cancer-associated genes was synthesized. Serum samples from healthy male volunteers (n=15) and patients with prostate cancer (n=85) were used to probe the array. These samples included patients with various clinical stages of disease: newly diagnosed localized prostate cancer (n=15), castration-sensitive non-metastatic prostate cancer (nmCSPC, n=40), castration-resistant non-metastatic prostate cancer (n=15) and castration-resistant metastatic disease (n=15). The patients with nmCSPC received treatment with either standard androgen deprivation therapy (ADT) or an antitumor DNA vaccine encoding prostatic acid phosphatase. Serial sera samples from these individuals were also used to probe the array, to secondarily determine whether this approach could be used to detect treatment-related changes.ResultsWe demonstrated that this peptide array yielded highly reproducible measurements of serum IgG levels. We found that the overall number of antibody responses did not increase with disease burden. However, the composition of recognized proteins shifted with clinical stage of disease. Our analysis revealed that the largest difference was between patients with castration-sensitive and castration-resistant disease. Patients with castration-resistant disease recognized more proteins associated with nucleic acid binding and gene regulation compared with men in other groups. Our longitudinal data showed that treatments can elicit antibodies detectable by this array, and notably vaccine-treated patients developed increased responses to more proteins over the course of treatment than did ADT-treated patients.ConclusionsThis study represents the largest survey of prostate cancer-associated antibodies to date. We have been able to characterize the classes of proteins recognized by patients and determine how they change with disease burden. Our findings further demonstrate the potential of this platform for measuring antigen spread and studying responses to immunomodulatory therapies.
- Published
- 2020
23. Inducible expression of cancer-testis antigens in human prostate cancer
- Author
-
Stephanie M. Thiede, Erika Heninger, Madison R. Kircher, David Kosoff, Timothy E. G. Krueger, Jamie M. Sperger, Brianna L. Byers, Joshua M. Lang, David F. Jarrard, Douglas G. McNeel, and Bing Yang
- Subjects
Male ,0301 basic medicine ,Indoles ,medicine.medical_treatment ,Antineoplastic Agents ,Decitabine ,Hydroxamic Acids ,cancer testis antigen ,tumor immunotherapy ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Circulating tumor cell ,Immune system ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,Panobinostat ,Testis ,Biomarkers, Tumor ,medicine ,Humans ,epigenetics ,business.industry ,Membrane Proteins ,Prostatic Neoplasms ,Cancer ,Immunotherapy ,Neoplastic Cells, Circulating ,prostate cancer ,medicine.disease ,Neoplasm Proteins ,3. Good health ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,030104 developmental biology ,Oncology ,Hypomethylating agent ,030220 oncology & carcinogenesis ,Immunology ,Azacitidine ,Cancer research ,Cancer/testis antigens ,methylation ,business ,Research Paper - Abstract
// Erika Heninger 1, 3 , Timothy E.G. Krueger 3 , Stephanie M. Thiede 1, 3 , Jamie M. Sperger 1, 3 , Brianna L. Byers 3 , Madison R. Kircher 3 , David Kosoff 1, 3 , Bing Yang 2, 3 , David F. Jarrard 2, 3 , Douglas G. McNeel 1, 3 , Joshua M. Lang 1, 3 1 Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA 2 Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA 3 University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA Correspondence to: Joshua M. Lang, email: jmlang@medicine.wisc.edu Keywords: cancer testis antigen, prostate cancer, epigenetics, tumor immunotherapy, methylation Received: April 22, 2016 Accepted: October 11, 2016 Published: October 17, 2016 ABSTRACT Immune tolerance to self-antigens can limit robust anti-tumor immune responses in the use of tumor vaccines. Expression of novel tumor associated antigens can improve immune recognition and lysis of tumor cells. The cancer-testis antigen (CTA) family of proteins has been hypothesized to be an ideal class of antigens due to tumor-restricted expression, a subset of which have been found to induce antibody responses in patients with prostate disease. We demonstrate that CTA expression is highly inducible in five different Prostate Cancer (PC) cell lines using a hypomethylating agent 5-Aza-2′-deoxycytidine (5AZA) and/or a histone deacetylase inhibitor LBH589. These CTAs include NY-ESO1, multiple members of the MAGE and SSX families and NY-SAR35. A subset of CTAs is synergistically induced by the combination of 5AZA and LBH589. We developed an ex vivo organ culture using human PC biopsies for ex vivo drug treatments to evaluate these agents in clinical samples. These assays found significant induction of SSX2 in 9/9 distinct patient samples and NY-SAR35 in 7/9 samples. Further, we identify expression of SSX2 in circulating tumor cells (CTC) from patients with advanced PC. These results indicate that epigenetic modifying agents can induce expression of a broad range of neoantigens in human PC and may serve as a useful adjunctive therapy with novel tumor vaccines and checkpoint inhibitors.
- Published
- 2016
24. B lymphocytes as direct antigen-presenting cells for anti-tumor DNA vaccines
- Author
-
Viswa Teja Colluru and Douglas G. McNeel
- Subjects
CD4-Positive T-Lymphocytes ,DNA vaccine ,0301 basic medicine ,Antigen-Presenting Cells ,CD8-Positive T-Lymphocytes ,Biology ,Cancer Vaccines ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Plasmid ,Antigen ,Cell Line, Tumor ,Vaccines, DNA ,Animals ,Humans ,Cytotoxic T cell ,Immune response ,Antigen-presenting cell ,Mice, Knockout ,Antigen Presentation ,B-Lymphocytes ,B cells ,Immunogenicity ,Research Paper: Immunology ,Immunity ,Cross-presentation ,DNA ,Dendritic Cells ,Virology ,Coculture Techniques ,3. Good health ,Mice, Inbred C57BL ,direct presentation ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Immunology and Microbiology Section ,Sarcoma, Experimental ,Plasmids - Abstract
// Viswa Teja Colluru 1,2 and Douglas G. McNeel 1,2 1 Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA 2 Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA Correspondence to: Douglas G. McNeel, email: // Keywords : DNA vaccine, B cells, dendritic cells, direct presentation, Immunology and Microbiology Section, Immune response, Immunity Received : July 26, 2016 Accepted : September 16, 2016 Published : September 21, 2016 Abstract In spite of remarkable preclinical efficacy, DNA vaccination has demonstrated low immunogenicity in humans. While efforts have focused on increasing cross-presentation of DNA-encoded antigens, efforts to increase DNA vaccine immunogenicity by targeting direct presentation have remained mostly unexplored. In these studies, we compared the ability of different APCs to present antigen to T cells after simple co-culture with plasmid DNA. We found that human primary peripheral B lymphocytes, and not monocytes or in vitro derived dendritic cells (DCs), were able to efficiently encode antigen mRNA and expand cognate tumor antigen-specific CD8 T cells ex vivo . Similarly, murine B lymphocytes co-cultured with plasmid DNA, and not DCs, were able to prime antigen-specific T cells in vivo. Moreover, B lymphocyte-mediated presentation of plasmid antigen led to greater Th1-biased immunity and was sufficient to elicit an anti-tumor effect in vivo . Surprisingly, increasing plasmid presentation by B cells, and not cross presentation of peptides by DCs, further augmented traditional plasmid vaccination. Together, these data suggest that targeting plasmid DNA to B lymphocytes, for example through transfer of ex vivo plasmidloaded B cells, may be novel means to achieve greater T cell immunity from DNA vaccines.
- Published
- 2016
25. Immunotherapy for prostate cancer: False promises or true hope?
- Author
-
Douglas G. McNeel and Brian T. Rekoske
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Immune regulation ,Cancer ,Disease ,Immunotherapy ,medicine.disease ,Food and drug administration ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Immunology ,Advanced disease ,Medicine ,business - Abstract
Prostate cancer is the most commonly diagnosed cancer, and the second leading cause of cancer-related death for men in the United States. Despite the approval of several new agents for advanced disease, each of these has prolonged survival by only a few months. Consequently, new therapies are sorely needed. For other cancer types, immunotherapy has demonstrated dramatic and durable treatment responses, causing many to hope that immunotherapies might provide an ideal treatment approach for patients with advanced prostate cancer. However, apart from sipuleucel-T, prostate cancer has been conspicuously absent from the list of malignancies for which immunotherapies have received recent approval from the US Food and Drug Administration. This has left some wondering whether immunotherapy will "work" for this disease. In this review, the authors describe current developments in immunotherapy, including approaches to engage tumor-targeting T cells, disrupt immune regulation, and alter the immunosuppressive tumor microenvironment. The authors then describe the recent application of these approaches to the treatment of prostate cancer. Given the Food and Drug Administration approval of 1 agent, and the finding that several others are in advanced stages of clinical testing, the authors believe that immunotherapies offer real hope to improve patient outcomes for men with prostate cancer, especially as investigators begin to explore rational combinations of immunotherapies and combine these therapies with other conventional therapies. Cancer 2016;122:3598-607. © 2016 American Cancer Society.
- Published
- 2016
26. PD-1 or PD-L1 Blockade Restores Antitumor Efficacy Following SSX2 Epitope–Modified DNA Vaccine Immunization
- Author
-
Brett B. Maricque, Brian M. Olson, Brian T. Rekoske, Heath A. Smith, and Douglas G. McNeel
- Subjects
Cancer Research ,Programmed Cell Death 1 Receptor ,Immunology ,Gene Expression ,CD8-Positive T-Lymphocytes ,Cancer Vaccines ,B7-H1 Antigen ,Article ,Epitope ,DNA vaccination ,Epitopes ,Mice ,Antigen ,Cell Line, Tumor ,Neoplasms ,MHC class I ,Vaccines, DNA ,Animals ,Humans ,Mice, Knockout ,biology ,Immunogenicity ,Antibodies, Monoclonal ,Peptide Fragments ,Disease Models, Animal ,Immunization ,Cell culture ,biology.protein ,Cancer research ,CD8 - Abstract
DNA vaccines have demonstrated antitumor efficacy in multiple preclinical models, but low immunogenicity has been observed in several human clinical trials. This has led to many approaches seeking to improve the immunogenicity of DNA vaccines. We previously reported that a DNA vaccine encoding the cancer–testis antigen SSX2, modified to encode altered epitopes with increased MHC class I affinity, elicited a greater frequency of cytolytic, multifunctional CD8+ T cells in non–tumor-bearing mice. We sought to test whether this optimized vaccine resulted in increased antitumor activity in mice bearing an HLA-A2–expressing tumor engineered to express SSX2. We found that immunization of tumor-bearing mice with the optimized vaccine elicited a surprisingly inferior antitumor effect relative to the native vaccine. Both native and optimized vaccines led to increased expression of PD-L1 on tumor cells, but antigen-specific CD8+ T cells from mice immunized with the optimized construct expressed higher PD-1. Splenocytes from immunized animals induced PD-L1 expression on tumor cells in vitro. Antitumor activity of the optimized vaccine could be increased when combined with antibodies blocking PD-1 or PD-L1, or by targeting a tumor line not expressing PD-L1. These findings suggest that vaccines aimed at eliciting effector CD8+ T cells, and DNA vaccines in particular, might best be combined with PD-1 pathway inhibitors in clinical trials. This strategy may be particularly advantageous for vaccines targeting prostate cancer, a disease for which antitumor vaccines have demonstrated clinical benefit and yet PD-1 pathway inhibitors alone have shown little efficacy to date. Cancer Immunol Res; 3(8); 946–55. ©2015 AACR.
- Published
- 2015
27. Randomized phase II trial of docetaxel with or without PSA-TRICOM vaccine in patients with castrate-resistant metastatic prostate cancer: A trial of the ECOG-ACRIN cancer research group (E1809)
- Author
-
Douglas G. McNeel, Yu-Hui Chen, Ravi A. Madan, Robert S. DiPaola, Alexander J. Dwyer, Michael A. Carducci, and James L. Gulley
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Fowlpox ,Oncology ,medicine.medical_specialty ,Antibodies, Neoplasm ,medicine.medical_treatment ,Immunology ,Antineoplastic Agents ,Docetaxel ,CD8-Positive T-Lymphocytes ,Cancer Vaccines ,Prostate cancer ,Internal medicine ,Clinical endpoint ,Humans ,Immunology and Allergy ,Medicine ,Neoplasm Metastasis ,Survival analysis ,Aged ,Pharmacology ,Chemotherapy ,business.industry ,Middle Aged ,Prostate-Specific Antigen ,medicine.disease ,Survival Analysis ,Research Papers ,Clinical trial ,Prostatic Neoplasms, Castration-Resistant ,Prostate-specific antigen ,Treatment Outcome ,Immunoglobulin G ,Taxoids ,business ,medicine.drug - Abstract
Anti-tumor vaccines have demonstrated efficacy in patients with castration-resistant metastatic prostate cancer. One vaccine, Prostvac-VF®, using a heterologous prime-boost strategy with vaccinia and fowlpox viral vectors encoding PSA, is currently being evaluated in a registration phase III multinational clinical trial. The current trial was planned to assess the clinical efficacy of this vaccine in patients with castration-resistant metastatic prostate cancer receiving subsequent docetaxel chemotherapy. 10 patients with metastatic castration-resistant prostate cancer, with a predicted survival of at least 18 months, were enrolled out of a planned 144 patients. Eight of 10 patients were treated and were randomized to receive docetaxel chemotherapy alone (Arm B, n = 2) versus treatment with Prostvac-VF (days 1, 15, 29, 43, 57) followed by docetaxel (Arm A, n = 6) chemotherapy beginning at month 3. The primary endpoint of the trial was overall survival, and secondary endpoints included time to radiographic progression and immunological response. The trial was opened within the Eastern Cooperative Oncology Group, but due to slow accrual was closed by CTEP after only 10 patients were enrolled within 13 months. Results: Presented here are the safety, clinical, and immunological results from 8 eligible patients who underwent treatment. Two of 6 patients treated on Arm A, with vaccine followed by docetaxel, had a >50% PSA response, with one of these patients experiencing a PSA decline during treatment with vaccine. Significant PSA-specific CD4+ and CD8+ T-cell responses and IgG antibody responses specific for PSA were not detected. The primary endpoint of overall survival cannot be assessed due to limited accrual. The lack of T-cell responses, even in this small cohort, suggests that further validation and development of immune biomarkers will be important for future studies. Other trials remain ongoing to evaluate the role of anti-tumor vaccination in sequence with other traditional anti-tumor therapies.
- Published
- 2015
28. Pretreatment antigen-specific immunity and regulation - association with subsequent immune response to anti-tumor DNA vaccination
- Author
-
Brian M. Olson, Douglas G. McNeel, and Laura E. Johnson
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Prostatic acid phosphatase ,Mice, SCID ,Leukocyte Count ,Immunogenicity, Vaccine ,Interleukin 10 ,0302 clinical medicine ,Vaccines, DNA ,Immunology and Allergy ,Medicine ,Immunity, Cellular ,Prostate cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Interleukin-10 ,3. Good health ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Research Article ,DNA vaccine ,T cell ,Acid Phosphatase ,Immunology ,lcsh:RC254-282 ,Cancer Vaccines ,Immunophenotyping ,DNA vaccination ,Interferon-gamma ,03 medical and health sciences ,Immune system ,Antigen ,Antigens, Neoplasm ,Immunity ,Biomarkers, Tumor ,Animals ,Humans ,B cell ,Pharmacology ,business.industry ,Prostatic Neoplasms ,Biomarker ,030104 developmental biology ,Leukocytes, Mononuclear ,Myeloid-derived Suppressor Cell ,Cytokine secretion ,business - Abstract
Background Immunotherapies have demonstrated clinical benefit for many types of cancers, however many patients do not respond, and treatment-related adverse effects can be severe. Hence many efforts are underway to identify treatment predictive biomarkers. We have reported the results of two phase I trials using a DNA vaccine encoding prostatic acid phosphatase (PAP) in patients with biochemically recurrent prostate cancer. In both trials, persistent PAP-specific Th1 immunity developed in some patients, and this was associated with favorable changes in serum PSA kinetics. In the current study, we sought to determine if measures of antigen-specific or antigen non-specific immunity were present prior to treatment, and associated with subsequent immune response, to identify possible predictive immune biomarkers. Methods Patients who developed persistent PAP-specific, IFNγ-secreting immune responses were defined as immune “responders.” The frequency of peripheral T cell and B cell lymphocytes, natural killer cells, monocytes, dendritic cells, myeloid derived suppressor cells, and regulatory T cells were assessed by flow cytometry and clinical laboratory values. PAP-specific immune responses were evaluated by cytokine secretion in vitro, and by antigen-specific suppression of delayed-type hypersensitivity to a recall antigen in an in vivo SCID mouse model. Results The frequency of peripheral blood cell types did not differ between the immune responder and non-responder groups. Non-responder patients tended to have higher PAP-specific IL-10 production pre-vaccination (p = 0.09). Responder patients had greater preexisting PAP-specific bystander regulatory responses that suppressed DTH to a recall antigen (p = 0.016). Conclusions While our study population was small (n = 38), these results suggest that different measures of antigen-specific tolerance or regulation might help predict immunological outcome from DNA vaccination. These will be prospectively evaluated in an ongoing randomized, phase II trial. Electronic supplementary material The online version of this article (doi:10.1186/s40425-017-0260-3) contains supplementary material, which is available to authorized users.
- Published
- 2017
29. Vaccination with High-Affinity Epitopes Impairs Antitumor Efficacy by Increasing PD-1 Expression on CD8+ T Cells
- Author
-
Douglas G. McNeel, Christopher D. Zahm, and Viswa Teja Colluru
- Subjects
0301 basic medicine ,Cancer Research ,Adoptive cell transfer ,Immunology ,Programmed Cell Death 1 Receptor ,Receptors, Antigen, T-Cell ,Biology ,CD8-Positive T-Lymphocytes ,Major histocompatibility complex ,Cancer Vaccines ,Epitope ,Article ,B7-H1 Antigen ,Major Histocompatibility Complex ,03 medical and health sciences ,Epitopes ,Mice ,0302 clinical medicine ,Antigen ,Neoplasms ,Cytotoxic T cell ,Animals ,Humans ,Immunogenicity ,T-cell receptor ,Vaccination ,Adoptive Transfer ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Immunization ,biology.protein ,030215 immunology - Abstract
Antitumor vaccines encoding self-antigens generally have low immunogenicity in clinical trials. Several approaches are aimed at improving vaccine immunogenicity, including efforts to alter encoded epitopes. Immunization with epitopes altered for increased affinity for the major histocompatibility complex (MHC) or T-cell receptor (TCR) elicits greater numbers of CD8 T cells but inferior antitumor responses. Our previous results suggested that programmed death 1 (PD-1) and its ligand (PD-L1) increased on antigen-specific CD8 T cells and tumor cells, respectively, after high-affinity vaccination. In this report, we use two murine models to investigate whether the dose, MHC affinity, or TCR affinity of an epitope affected the antitumor response via the PD-1/PD-L1 axis. T cells activated with high-affinity epitopes resulted in prolonged APC:T-cell contact time that led to elevated, persistent PD-1 expression, and expression of other checkpoint molecules, in vitro and in vivo. Immunization with high-affinity epitopes also decreased antitumor efficacy in the absence of PD-1 blockade. Thus, APC:T-cell contact time can be altered by epitope affinity and lead to therapeutically relevant changes in vaccine efficacy mediated by changes in PD-1 expression. These findings have implications for the use of agents targeting PD-1 expression or function whenever high-affinity CD8 T cells are elicited or supplied by means of vaccination or adoptive transfer. Cancer Immunol Res; 5(8); 630–41. ©2017 AACR.
- Published
- 2017
30. Safety and Immunological Efficacy of a DNA Vaccine Encoding the Androgen Receptor Ligand-Binding Domain (AR-LBD)
- Author
-
Jens C. Eickhoff, Douglas G. McNeel, Brian M. Olson, Jordan E. Bloom, Eric S. Bradley, Thomas Sawicki, Erik A. Ranheim, Laura E. Johnson, Brian T. Rekoske, Viswa Teja Colluru, and Weixiong Zhong
- Subjects
Male ,Urology ,Article ,DNA vaccination ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,Immune system ,Adjuvants, Immunologic ,Prostate ,Immunity ,Monitoring, Immunologic ,medicine ,Vaccines, DNA ,Animals ,business.industry ,Granulocyte-Macrophage Colony-Stimulating Factor ,Prostatic Neoplasms ,medicine.disease ,Androgen receptor ,Vaccination ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Immunology ,business ,CD8 ,030215 immunology - Abstract
BACKGROUND The androgen receptor (AR) is a key oncogenic driver of prostate cancer, and has been the primary focus of prostate cancer treatment for several decades. We have previously demonstrated that the AR is also an immunological target antigen, recognized in patients with prostate cancer, and targetable by means of vaccines in rodent models with delays in prostate tumor growth. The current study was performed to determine the safety and immunological efficacy of a GMP-grade plasmid DNA vaccine encoding the ligand-binding domain (LBD) of the AR, pTVG-AR. METHODS Groups of male mice (n = 6–10 per group) were evaluated after four or seven immunizations, using different schedules and inclusion of GM-CSF as a vaccine adjuvant. Animals were assessed for toxicity using gross observations, pathological analysis, and analysis of serum chemistries. Animals were analyzed for evidence of vaccine-augmented immunity by tetramer analysis. Survival studies using different immunization schedules and inclusion of GM-CSF were conducted in an autochthonous genetically engineered mouse model. RESULTS No significant toxicities were observed in terms of animal weights, histopathology, hematological changes, or changes in serum chemistries, although there was a trend to lower serum glucose in animals treated with the vaccine. There was specifically no evidence of toxicity in other tissues that express AR, including liver, muscle, hematopoietic, and brain. Vaccination was found to elicit AR LBD-specific CD8+ T cells. In a subsequent study of tumor-bearing animals, animals treated with vaccine had prolonged survival compared with control-immunized mice. CONCLUSIONS These studies demonstrate that, in immunocompetent mice expressing the target antigen, immunization with the pTVG-AR vaccine was both safe and effective in eliciting AR-specific cellular immune responses, and prolonged the survival of prostate tumor-bearing mice. These findings support the clinical evaluation of pTVG-AR in patients with recurrent prostate cancer. Prostate 77:812–821, 2017. © 2017 Wiley Periodicals, Inc.
- Published
- 2017
31. A Transient Increase in Eosinophils Is Associated with Prolonged Survival in Men with Metastatic Castration-Resistant Prostate Cancer Who Receive Sipuleucel-T
- Author
-
Robert B. Sims, Mark H. Wener, Douglas G. McNeel, Philip W. Kantoff, Robert Dreicer, Celestia S. Higano, Thomas A. Gardner, Eric J. Small, Todd DeVries, and Nadeem A. Sheikh
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_treatment ,Phases of clinical research ,Kaplan-Meier Estimate ,Castration-Resistant ,Leukocyte Count ,Prostate cancer ,Prostate ,80 and over ,Cancer ,Aged, 80 and over ,Prostate Cancer ,Blood Proteins ,Pharmacology and Pharmaceutical Sciences ,Middle Aged ,Prognosis ,Prostatic Neoplasms, Castration-Resistant ,Sipuleucel-T ,Treatment Outcome ,medicine.anatomical_structure ,6.1 Pharmaceuticals ,medicine.symptom ,medicine.drug ,Adult ,Urologic Diseases ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Immunology ,Bone Neoplasms ,Cancer Vaccines ,Asymptomatic ,Article ,Double-Blind Method ,Clinical Research ,Internal medicine ,medicine ,Humans ,Retrospective Studies ,Aged ,Chemotherapy ,Tissue Extracts ,business.industry ,Prostatic Neoplasms ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Eosinophils ,Clinical trial ,business - Abstract
Sipuleucel-T is an autologous cellular immunotherapy used to treat asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC). Traditional short-term indicators of clinical response commonly used with chemotherapy have not correlated with survival in patients treated with sipuleucel-T. This retrospective study aimed to evaluate laboratory parameters as possible early biomarkers associated with clinical benefit following sipuleucel-T treatment. Patients treated with sipuleucel-T from three randomized, controlled, phase III clinical trials in mCRPC were considered: IMPACT (NCT00065442; n = 512), D9901 (NCT00005947; n = 127), and D9902A (NCT01133704; n = 98). Patients from these trials were included in this study if their samples were analyzed by the central laboratory and if data were available from baseline and ≥1 posttreatment time point (n = 377). We found that sipuleucel-T treatment was associated with a transient increase in serum eosinophil count at week 6 that resolved by week 14 in 28% of patients (105 of 377). This eosinophil increase correlated with induced immune response, longer prostate cancer–specific survival [HR, 0.713; 95% confidence interval (CI), 0.525–0.970; P = 0.031], and a trend in overall survival (HR, 0.753; 95% CI, 0.563–1.008; P = 0.057). Median serum globulin protein levels also increased transiently, which was associated with antigen-specific antibody responses; however, this finding did not correlate with longer survival. We conclude that transient increases in eosinophils at week 6 may be a useful, objective, short-term indicator of global immune activation and survival benefit with sipuleucel-T in patients with mCRPC. This observation warrants prospective evaluation in future clinical trials. Cancer Immunol Res; 2(10); 988–99. ©2014 AACR.
- Published
- 2014
32. DNA vaccines encoding altered peptide ligands for SSX2 enhance epitope-specific CD8+ T-cell immune responses
- Author
-
Brian T. Rekoske, Douglas G. McNeel, and Heath A. Smith
- Subjects
Male ,Epitopes, T-Lymphocyte ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Biology ,Ligands ,Major histocompatibility complex ,Cancer Vaccines ,Article ,Epitope ,DNA vaccination ,Mice ,Immune system ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,HLA-A2 Antigen ,Vaccines, DNA ,Animals ,Humans ,Cytotoxic T cell ,Mice, Knockout ,General Veterinary ,General Immunology and Microbiology ,Immunodominant Epitopes ,Immunogenicity ,Public Health, Environmental and Occupational Health ,Prostatic Neoplasms ,Virology ,Peptide Fragments ,Neoplasm Proteins ,Mice, Inbred C57BL ,Repressor Proteins ,Infectious Diseases ,Immunology ,biology.protein ,Molecular Medicine - Abstract
Plasmid DNA serves as a simple and easily modifiable form of antigen delivery for vaccines. The USDA approval of DNA vaccines for several non-human diseases underscores the potential of this type of antigen delivery method as a cost-effective approach for the treatment or prevention of human diseases, including cancer. However, while DNA vaccines have demonstrated safety and immunological effect in early phase clinical trials, they have not consistently elicited robust anti-tumor responses. Hence many recent efforts have sought to increase the immunological efficacy of DNA vaccines, and we have specifically evaluated several target antigens encoded by DNA vaccine as treatments for human prostate cancer. In particular, we have focused on SSX2 as one potential target antigen, given its frequent expression in metastatic prostate cancer. We have previously identified two peptides, p41-49 and p103-111, as HLA-A2-restricted SSX2-specific epitopes. In the present study we sought to determine whether the efficacy of a DNA vaccine could be enhanced by an altered peptide ligand (APL) strategy wherein modifications were made to anchor residues of these epitopes to enhance or ablate their binding to HLA-A2. A DNA vaccine encoding APL modified to increase epitope binding elicited robust peptide-specific CD8+ T cells producing Th1 cytokines specific for each epitope. Ablation of one epitope in a DNA vaccine did not enhance immune responses to the other epitope. These results demonstrate that APL encoded by a DNA vaccine can be used to elicit increased numbers of antigen-specific T cells specific for multiple epitopes simultaneously, and suggest this could be a general approach to improve the immunogenicity of DNA vaccines encoding tumor antigens.
- Published
- 2014
33. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of prostate carcinoma
- Author
-
Daniel P. Petrylak, Douglas G. McNeel, Oliver Sartor, Stacey Harrelson, David Peace, William Oh, James L. Gulley, Mark N. Stein, Neal D. Shore, Charles G. Drake, Philip W. Kantoff, Lawrence Fong, Susan F. Slovin, Tomasz M. Beer, Ravi A. Madan, Johannes Vieweg, Hank Porterfield, and Neil H. Bander
- Subjects
0301 basic medicine ,Oncology ,Urologic Diseases ,Cancer Research ,medicine.medical_specialty ,Aging ,medicine.medical_treatment ,Immunology ,Castrate-resistant prostate cancer ,Guidelines ,Malignancy ,Vaccine Related ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Internal medicine ,Position Article and Guidelines ,medicine ,Immunology and Allergy ,Cancer ,Pharmacology ,business.industry ,Prostate Cancer ,Immunotherapeutic agent ,Prostate carcinoma ,Immunotherapy ,medicine.disease ,Clinical trial ,Treatment ,030104 developmental biology ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Molecular Medicine ,Immunization ,Development of treatments and therapeutic interventions ,business ,Biotechnology - Abstract
Prostate cancer is the most commonly diagnosed malignancy and second leading cause of cancer death among men in the United States. In recent years, several new agents, including cancer immunotherapies, have been approved or are currently being investigated in late-stage clinical trials for the management of advanced prostate cancer. Therefore, the Society for Immunotherapy of Cancer (SITC) convened a multidisciplinary panel, including physicians, nurses, and patient advocates, to develop consensus recommendations for the clinical application of immunotherapy for prostate cancer patients. To do so, a systematic literature search was performed to identify high-impact papers from 2006 until 2014 and was further supplemented with literature provided by the panel. Results from the consensus panel voting and discussion as well as the literature review were used to rate supporting evidence and generate recommendations for the use of immunotherapy in prostate cancer patients. Sipuleucel-T, an autologous dendritic cell vaccine, is the first and currently only immunotherapeutic agent approved for the clinical management of metastatic castrate resistant prostate cancer (mCRPC). The consensus panel utilized this model to discuss immunotherapy in the treatment of prostate cancer, issues related to patient selection, monitoring of patients during and post treatment, and sequence/combination with other anti-cancer treatments. Potential immunotherapies emerging from late-stage clinical trials are also discussed. As immunotherapy evolves as a therapeutic option for the treatment of prostate cancer, these recommendations will be updated accordingly. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0198-x) contains supplementary material, which is available to authorized users.
- Published
- 2016
34. A negative selection methodology using a microfluidic platform for the isolation and enumeration of circulating tumor cells
- Author
-
David J. Beebe, Scott M. Berry, Jordan T. Becker, Joshua M. Lang, Jay W. Warrick, Rachel Mosher, Lindsey J. Maccoux, Benjamin P. Casavant, Vivian Chen, and Douglas G. McNeel
- Subjects
Male ,Enzyme-Linked Immunospot Assay ,Cell ,Population ,Cell Count ,Cell Separation ,Computational biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Negative selection ,chemistry.chemical_compound ,Circulating tumor cell ,Antigens, Neoplasm ,Neoplasms ,medicine ,Enumeration ,Humans ,Neoplasm Metastasis ,education ,Molecular Biology ,education.field_of_study ,biology ,ELISPOT ,Epithelial cell adhesion molecule ,Microfluidic Analytical Techniques ,Epithelial Cell Adhesion Molecule ,Neoplastic Cells, Circulating ,medicine.anatomical_structure ,chemistry ,Immunology ,biology.protein ,Antibody ,Cell Adhesion Molecules - Abstract
Circulating tumor cells (CTCs) exist in the peripheral blood stream of metastatic cancer patients at rates of approximately 1 CTC per billion background cells. In order to capture and analyze this rare cell popu- lation, various techniques exist that range from antibody-based surface marker positive selection to methods that use physical properties of CTCs to negatively exclude background cells from a CTC popula- tion. However, methods to capture cells for functional downstream analyses are limited due to inacces- sibility of the captured sample or labeling techniques that may be prohibitive to cell function. Here, we present a negative selection method that leverages a Microfluidic Cell Concentrator (MCC) to allow col- lection and analysis of this rare cell population without needing cell adhesion or other labeling tech- niques to keep the cells within the chamber. Because the MCC is designed to allow collection and analysis of non-adherent cell populations, multiple staining steps can be applied in parallel to a given CTC population without losing any of the population. The ability of the MCC for patient sample processing of CTCs for enumeration was demonstrated with five patient samples, revealing an average of 0.31 CTCs / mL. The technique was compared to a previously published method - the ELISPOT - that showed similar CTC levels among the five patient samples tested. Because the MCC method does not use positive selec- tion, the method can be applied across a variety of tumor types with no changes to the process.
- Published
- 2013
35. Vaccination using peptides spanning the SYT–SSX tumor-specific translocation
- Author
-
Jordan E. Bloom, Brian M. Olson, and Douglas G. McNeel
- Subjects
Pharmacology ,business.industry ,Immunology ,Breakpoint ,Cancer ,Chromosomal translocation ,medicine.disease ,Synovial sarcoma ,Vaccination ,Drug Discovery ,Peptide vaccine ,medicine ,Cancer research ,Molecular Medicine ,Sarcoma ,business ,Chronic myelogenous leukemia - Abstract
Evaluation of: Kawaguchi S, Tsukahara T, Ida K et al. SYT–SSX breakpoint peptide vaccines in patients with synovial sarcoma: a study from the Japanese Musculoskeletal Oncology Group. Cancer Sci. 103(9), 1625–1630 (2012).The identification of genetic translocations as key tumor-initiating events has led to the development of novel antigen-specific vaccines targeting these tumor-specific breakpoint regions. Previous studies have evaluated vaccines targeting the breakpoints in the BCR-ABL translocation in patients with chronic myelogenous leukemia and EWS-FLI1 in patients with Ewing sarcoma. In the article under evaluation, the authors evaluated a peptide vaccine targeting the breakpoint in the SYT–SSX translocation, the genetic translocation essentially pathognomonic for synovial sarcoma. This is the second small clinical trial reported by this group using HLA-A24-binding peptides as vaccine antigens. In this four-arm trial, using a native or HLA-A24-optimized SYT–SSX peptide with or without adjuvant plus I...
- Published
- 2012
36. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): part two
- Author
-
Casey Ager, Matthew Reilley, Courtney Nicholas, Todd Bartkowiak, Ashvin Jaiswal, Michael Curran, Tina C. Albershardt, Anshika Bajaj, Jacob F. Archer, Rebecca S. Reeves, Lisa Y. Ngo, Peter Berglund, Jan ter Meulen, Caroline Denis, Hormas Ghadially, Thomas Arnoux, Fabien Chanuc, Nicolas Fuseri, Robert W. Wilkinson, Nicolai Wagtmann, Yannis Morel, Pascale Andre, Michael B. Atkins, Matteo S. Carlino, Antoni Ribas, John A. Thompson, Toni K. Choueiri, F. Stephen Hodi, Wen-Jen Hwu, David F. McDermott, Victoria Atkinson, Jonathan S. Cebon, Bernie Fitzharris, Michael B. Jameson, Catriona McNeil, Andrew G. Hill, Eric Mangin, Malidi Ahamadi, Marianne van Vugt, Mariëlle van Zutphen, Nageatte Ibrahim, Georgina V. Long, Robyn Gartrell, Zoe Blake, Ines Simoes, Yichun Fu, Takuro Saito, Yingzhi Qian, Yan Lu, Yvonne M. Saenger, Sadna Budhu, Olivier De Henau, Roberta Zappasodi, Kyle Schlunegger, Bruce Freimark, Jeff Hutchins, Christopher A. Barker, Jedd D. Wolchok, Taha Merghoub, Elena Burova, Omaira Allbritton, Peter Hong, Jie Dai, Jerry Pei, Matt Liu, Joel Kantrowitz, Venus Lai, William Poueymirou, Douglas MacDonald, Ella Ioffe, Markus Mohrs, William Olson, Gavin Thurston, Cristian Capasso, Federica Frascaro, Sara Carpi, Siri Tähtinen, Sara Feola, Manlio Fusciello, Karita Peltonen, Beatriz Martins, Madeleine Sjöberg, Sari Pesonen, Tuuli Ranki, Lukasz Kyruk, Erkko Ylösmäki, Vincenzo Cerullo, Fabio Cerignoli, Biao Xi, Garret Guenther, Naichen Yu, Lincoln Muir, Leyna Zhao, Yama Abassi, Víctor Cervera-Carrascón, Mikko Siurala, João Santos, Riikka Havunen, Suvi Parviainen, Akseli Hemminki, Angus Dalgleish, Satvinder Mudan, Mark DeBenedette, Ana Plachco, Alicia Gamble, Elizabeth W. Grogan, John Krisko, Irina Tcherepanova, Charles Nicolette, Pooja Dhupkar, Ling Yu, Eugenie S. Kleinerman, Nancy Gordon, Italia Grenga, Lauren Lepone, Sofia Gameiro, Karin M. Knudson, Massimo Fantini, Kwong Tsang, James Hodge, Renee Donahue, Jeffrey Schlom, Elizabeth Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Cathie Foster, Alan Howell, Leslie Balch, Alyssa Knapp, John E. Leonard, Mark Paris, Terry Fisher, Siwen Hu-Lieskovan, Ernest Smith, Maurice Zauderer, William Fogler, Marilyn Franklin, Matt Thayer, Dan Saims, John L. Magnani, Jian Gong, Michael Gray, George Fromm, Suresh de Silva, Louise Giffin, Xin Xu, Jason Rose, Taylor H. Schreiber, Sofia R. Gameiro, Paul E. Clavijo, Clint T. Allen, James W. Hodge, Kwong Y. Tsang, Jane Grogan, Nicholas Manieri, Eugene Chiang, Patrick Caplazi, Mahesh Yadav, Patrick Hagner, Hsiling Chiu, Michelle Waldman, Anke Klippel, Anjan Thakurta, Michael Pourdehnad, Anita Gandhi, Ian Henrich, Laura Quick, Rob Young, Margaret Chou, Andrew Hotson, Stephen Willingham, Po Ho, Carmen Choy, Ginna Laport, Ian McCaffery, Richard Miller, Kimberly A. Tipton, Kenneth R. Wong, Victoria Singson, Chihunt Wong, Chanty Chan, Yuanhiu Huang, Shouchun Liu, Jennifer H. Richardson, W. Michael Kavanaugh, James West, Bryan A. Irving, Ritika Jaini, Matthew Loya, Charis Eng, Melissa L. Johnson, Alex A. Adjei, Mateusz Opyrchal, Suresh Ramalingam, Pasi A. Janne, George Dominguez, Dmitry Gabrilovich, Laura de Leon, Jeannette Hasapidis, Scott J. Diede, Peter Ordentlich, Scott Cruickshank, Michael L. Meyers, Matthew D. Hellmann, Pawel Kalinski, Amer Zureikat, Robert Edwards, Ravi Muthuswamy, Nataša Obermajer, Julie Urban, Lisa H. Butterfield, William Gooding, Herbert Zeh, David Bartlett, Olga Zubkova, Larissa Agapova, Marina Kapralova, Liudmila Krasovskaia, Armen Ovsepyan, Maxim Lykov, Artem Eremeev, Vladimir Bokovanov, Olga Grigoryeva, Andrey Karpov, Sergey Ruchko, Alexandr Shuster, Danny N. Khalil, Luis Felipe Campesato, Yanyun Li, Adam S. Lazorchak, Troy D. Patterson, Yueyun Ding, Pottayil Sasikumar, Naremaddepalli Sudarshan, Nagaraj Gowda, Raghuveer Ramachandra, Dodheri Samiulla, Sanjeev Giri, Rajesh Eswarappa, Murali Ramachandra, David Tuck, Timothy Wyant, Jasmin Leshem, Xiu-fen Liu, Tapan Bera, Masaki Terabe, Birgit Bossenmaier, Gerhard Niederfellner, Yoram Reiter, Ira Pastan, Leiming Xia, Yang Xia, Yangyang Hu, Yi Wang, Yangyi Bao, Fu Dai, Shiang Huang, Elaine Hurt, Robert E. Hollingsworth, Lawrence G. Lum, Alfred E. Chang, Max S. Wicha, Qiao Li, Thomas Mace, Neil Makhijani, Erin Talbert, Gregory Young, Denis Guttridge, Darwin Conwell, Gregory B. Lesinski, Rodney JM Macedo Gonzales, Austin P. Huffman, Ximi K. Wang, Ran Reshef, Andy MacKinnon, Jason Chen, Matt Gross, Gisele Marguier, Peter Shwonek, Natalija Sotirovska, Susanne Steggerda, Francesco Parlati, Amani Makkouk, Mark K. Bennett, Ethan Emberley, Tony Huang, Weiqun Li, Silinda Neou, Alison Pan, Jing Zhang, Winter Zhang, Netonia Marshall, Thomas U. Marron, Judith Agudo, Brian Brown, Joshua Brody, Christopher McQuinn, Matthew Farren, Hannah Komar, Reena Shakya, Thomas Ludwug, Y. Maurice Morillon, Scott A. Hammond, John W. Greiner, Pulak R. Nath, Anthony L. Schwartz, Dragan Maric, David D. Roberts, Aung Naing, Kyriakos P. Papadopoulos, Karen A. Autio, Deborah J. Wong, Manish Patel, Gerald Falchook, Shubham Pant, Patrick A. Ott, Melinda Whiteside, Amita Patnaik, John Mumm, Filip Janku, Ivan Chan, Todd Bauer, Rivka Colen, Peter VanVlasselaer, Gail L. Brown, Nizar M. Tannir, Martin Oft, Jeffrey Infante, Evan Lipson, Ajay Gopal, Sattva S. Neelapu, Philippe Armand, Stephen Spurgeon, John P. Leonard, Rachel E. Sanborn, Ignacio Melero, Thomas F. Gajewski, Matthew Maurer, Serena Perna, Andres A. Gutierrez, Raphael Clynes, Priyam Mitra, Satyendra Suryawanshi, Douglas Gladstone, Margaret K. Callahan, James Crooks, Sheila Brown, Audrey Gauthier, Marc Hillairet de Boisferon, Andrew MacDonald, Laura Rosa Brunet, William T. Rothwell, Peter Bell, James M. Wilson, Fumi Sato-Kaneko, Shiyin Yao, Shannon S. Zhang, Dennis A. Carson, Cristina Guiducci, Robert L. Coffman, Kazutaka Kitaura, Takaji Matsutani, Ryuji Suzuki, Tomoko Hayashi, Ezra E. W. Cohen, David Schaer, Yanxia Li, Julie Dobkin, Michael Amatulli, Gerald Hall, Thompson Doman, Jason Manro, Frank Charles Dorsey, Lillian Sams, Rikke Holmgaard, Krishnadatt Persaud, Dale Ludwig, David Surguladze, John S. Kauh, Ruslan Novosiadly, Michael Kalos, Kyla Driscoll, Hardev Pandha, Christy Ralph, Kevin Harrington, Brendan Curti, Wallace Akerley, Sumati Gupta, Alan Melcher, David Mansfield, David R. Kaufman, Emmett Schmidt, Mark Grose, Bronwyn Davies, Roberta Karpathy, Darren Shafren, Katerina Shamalov, Cyrille Cohen, Naveen Sharma, James Allison, Tala Shekarian, Sandrine Valsesia-Wittmann, Christophe Caux, Aurelien Marabelle, Brian M. Slomovitz, Kathleen M. Moore, Hagop Youssoufian, Marshall Posner, Poonam Tewary, Alan D. Brooks, Ya-Ming Xu, Kithsiri Wijeratne, Leslie A. A. Gunatilaka, Thomas J. Sayers, John P. Vasilakos, Tesha Alston, Simon Dovedi, James Elvecrog, Iwen Grigsby, Ronald Herbst, Karen Johnson, Craig Moeckly, Stefanie Mullins, Kristen Siebenaler, Julius SternJohn, Ashenafi Tilahun, Mark A. Tomai, Katharina Vogel, Eveline E. Vietsch, Anton Wellstein, Martin Wythes, Stefano Crosignani, Joseph Tumang, Shilpa Alekar, Patrick Bingham, Sandra Cauwenberghs, Jenny Chaplin, Deepak Dalvie, Sofie Denies, Coraline De Maeseneire, JunLi Feng, Kim Frederix, Samantha Greasley, Jie Guo, James Hardwick, Stephen Kaiser, Katti Jessen, Erick Kindt, Marie-Claire Letellier, Wenlin Li, Karen Maegley, Reece Marillier, Nichol Miller, Brion Murray, Romain Pirson, Julie Preillon, Virginie Rabolli, Chad Ray, Kevin Ryan, Stephanie Scales, Jay Srirangam, Jim Solowiej, Al Stewart, Nicole Streiner, Vince Torti, Konstantinos Tsaparikos, Xianxian Zheng, Gregory Driessens, Bruno Gomes, Manfred Kraus, Chunxiao Xu, Yanping Zhang, Giorgio Kradjian, Guozhong Qin, Jin Qi, Xiaomei Xu, Bo Marelli, Huakui Yu, Wilson Guzman, Rober Tighe, Rachel Salazar, Kin-Ming Lo, Jessie English, Laszlo Radvanyi, Yan Lan, Michael Postow, Yasin Senbabaoglu, Billel Gasmi, Hong Zhong, Cailian Liu, Daniel Hirschhorhn-Cymerman, Yuanyuan Zha, Gregory Malnassy, Noreen Fulton, Jae-Hyun Park, Wendy Stock, Yusuke Nakamura, Hongtao Liu, Xiaoming Ju, Rachelle Kosoff, Kimberly Ramos, Brandon Coder, Robert Petit, Michael Princiotta, Kyle Perry, Jun Zou, Ainhoa Arina, Christian Fernandez, Wenxin Zheng, Michael A. Beckett, Helena J. Mauceri, Yang-Xin Fu, Ralph R. Weichselbaum, Whitney Lewis, Yanyan Han, Yeting Wu, Chou Yang, Jing Huang, Dongyun Wu, Jin Li, Xiaoling Liang, Xiangjun Zhou, Jinlin Hou, Raffit Hassan, Thierry Jahan, Scott J. Antonia, Hedy L. Kindler, Evan W. Alley, Somayeh Honarmand, Weiqun Liu, Meredith L. Leong, Chan C. Whiting, Nitya Nair, Amanda Enstrom, Edward E. Lemmens, Takahiro Tsujikawa, Sushil Kumar, Lisa M. Coussens, Aimee L. Murphy, Dirk G. Brockstedt, Sven D. Koch, Martin Sebastian, Christian Weiss, Martin Früh, Miklos Pless, Richard Cathomas, Wolfgang Hilbe, Georg Pall, Thomas Wehler, Jürgen Alt, Helge Bischoff, Michael Geissler, Frank Griesinger, Jens Kollmeier, Alexandros Papachristofilou, Fatma Doener, Mariola Fotin-Mleczek, Madeleine Hipp, Henoch S. Hong, Karl-Josef Kallen, Ute Klinkhardt, Claudia Stosnach, Birgit Scheel, Andreas Schroeder, Tobias Seibel, Ulrike Gnad-Vogt, Alfred Zippelius, Ha-Ram Park, Yong-Oon Ahn, Tae Min Kim, Soyeon Kim, Seulki Kim, Yu Soo Lee, Bhumsuk Keam, Dong-Wan Kim, Dae Seog Heo, Shari Pilon-Thomas, Amy Weber, Jennifer Morse, Krithika Kodumudi, Hao Liu, John Mullinax, Amod A. Sarnaik, Luke Pike, Andrew Bang, Tracy Balboni, Allison Taylor, Alexander Spektor, Tyler Wilhite, Monica Krishnan, Daniel Cagney, Brian Alexander, Ayal Aizer, Elizabeth Buchbinder, Mark Awad, Leena Ghandi, Jonathan Schoenfeld, Elizabeth Lessey-Morillon, Lisa Ridnour, Neil H. Segal, Manish Sharma, Dung T. Le, Robert L. Ferris, Andrew D. Zelenetz, Ronald Levy, Izidore S. Lossos, Caron Jacobson, Radhakrishnan Ramchandren, John Godwin, A. Dimitrios Colevas, Roland Meier, Suba Krishnan, Xuemin Gu, Jaclyn Neely, John Timmerman, Claire I. Vanpouille-Box, Silvia C. Formenti, Sandra Demaria, Erik Wennerberg, Aranzazu Mediero, Bruce N. Cronstein, Michael P. Gustafson, AriCeli DiCostanzo, Courtney Wheatley, Chul-Ho Kim, Svetlana Bornschlegl, Dennis A. Gastineau, Bruce D. Johnson, Allan B. Dietz, Cameron MacDonald, Mark Bucsek, Guanxi Qiao, Bonnie Hylander, Elizabeth Repasky, William J. Turbitt, Yitong Xu, Andrea Mastro, Connie J. Rogers, Sita Withers, Ziming Wang, Lam T. Khuat, Cordelia Dunai, Bruce R. Blazar, Dan Longo, Robert Rebhun, Steven K. Grossenbacher, Arta Monjazeb, William J. Murphy, Scott Rowlinson, Giulia Agnello, Susan Alters, David Lowe, Nicole Scharping, Ashley V. Menk, Ryan Whetstone, Xue Zeng, Greg M. Delgoffe, Patricia M. Santos, Jian Shi, Greg Delgoffe, Misako Nagasaka, Ammar Sukari, Miranda Byrne-Steele, Wenjing Pan, Xiaohong Hou, Brittany Brown, Mary Eisenhower, Jian Han, Natalie Collins, Robert Manguso, Hans Pope, Yashaswi Shrestha, Jesse Boehm, W. Nicholas Haining, Kyle R. Cron, Ayelet Sivan, Keston Aquino-Michaels, Marco Orecchioni, Davide Bedognetti, Wouter Hendrickx, Claudia Fuoco, Filomena Spada, Francesco Sgarrella, Gianni Cesareni, Francesco Marincola, Kostas Kostarelos, Alberto Bianco, Lucia Delogu, Jessica Roelands, Sabri Boughorbel, Julie Decock, Scott Presnell, Ena Wang, Franco M. Marincola, Peter Kuppen, Michele Ceccarelli, Darawan Rinchai, Damien Chaussabel, Lance Miller, Andrew Nguyen, J. Zachary Sanborn, Charles Vaske, Shahrooz Rabizadeh, Kayvan Niazi, Steven Benz, Shashank Patel, Nicholas Restifo, James White, Sam Angiuoli, Mark Sausen, Sian Jones, Maria Sevdali, John Simmons, Victor Velculescu, Luis Diaz, Theresa Zhang, Jennifer S. Sims, Sunjay M. Barton, Angela Kadenhe-Chiweshe, Filemon Dela Cruz, Andrew T. Turk, Christopher F. Mazzeo, Andrew L. Kung, Jeffrey N. Bruce, Darrell J. Yamashiro, Eileen P. Connolly, Jason Baird, Marka Crittenden, David Friedman, Hong Xiao, Rom Leidner, Bryan Bell, Kristina Young, Michael Gough, Zhen Bian, Koby Kidder, Yuan Liu, Emily Curran, Xiufen Chen, Leticia P. Corrales, Justin Kline, Ethan G. Aguilar, Jennifer Guerriero, Alaba Sotayo, Holly Ponichtera, Alexandra Pourzia, Sara Schad, Ruben Carrasco, Suzan Lazo, Roderick Bronson, Anthony Letai, Richard S. Kornbluth, Sachin Gupta, James Termini, Elizabeth Guirado, Geoffrey W. Stone, Christina Meyer, Laura Helming, Nicholas Wilson, Robert Hofmeister, Natalie J. Neubert, Laure Tillé, David Barras, Charlotte Soneson, Petra Baumgaertner, Donata Rimoldi, David Gfeller, Mauro Delorenzi, Silvia A. Fuertes Marraco, Daniel E. Speiser, Tara S. Abraham, Bo Xiang, Michael S. Magee, Scott A. Waldman, Adam E. Snook, Wojciech Blogowski, Ewa Zuba-Surma, Marta Budkowska, Daria Salata, Barbara Dolegowska, Teresa Starzynska, Leo Chan, Srinivas Somanchi, Kelsey McCulley, Dean Lee, Nico Buettner, Feng Shi, Paisley T. Myers, Stuart Curbishley, Sarah A. Penny, Lora Steadman, David Millar, Ellen Speers, Nicola Ruth, Gabriel Wong, Robert Thimme, David Adams, Mark Cobbold, Remy Thomas, Mariam Al-Muftah, Michael KK Wong, Michael Morse, Joseph I. Clark, Howard L. Kaufman, Gregory A. Daniels, Hong Hua, Tharak Rao, Janice P. Dutcher, Kai Kang, Yogen Saunthararajah, Vamsidhar Velcheti, Vikas Kumar, Firoz Anwar, Amita Verma, Zinal Chheda, Gary Kohanbash, John Sidney, Kaori Okada, Shruti Shrivastav, Diego A. Carrera, Shuming Liu, Naznin Jahan, Sabine Mueller, Ian F. Pollack, Angel M. Carcaboso, Alessandro Sette, Yafei Hou, Hideho Okada, Jessica J. Field, Weiping Zeng, Vincent FS Shih, Che-Leung Law, Peter D. Senter, Shyra J. Gardai, Nicole M. Okeley, Jennifer G. Abelin, Abu Z. Saeed, Stacy A. Malaker, Jeffrey Shabanowitz, Stephen T. Ward, Donald F. Hunt, Pam Profusek, Laura Wood, Dale Shepard, Petros Grivas, Kerstin Kapp, Barbara Volz, Detlef Oswald, Burghardt Wittig, Manuel Schmidt, Julian P. Sefrin, Lars Hillringhaus, Valeria Lifke, Alexander Lifke, Anna Skaletskaya, Jose Ponte, Thomas Chittenden, Yulius Setiady, Eva Sivado, Vincent Thomas, Meddy El Alaoui, Sébastien Papot, Charles Dumontet, Mike Dyson, John McCafferty, Said El Alaoui, Praveen K. Bommareddy, Andrew Zloza, Frederick Kohlhapp, Ann W. Silk, Sachin Jhawar, Tomas Paneque, Jenna Newman, Pedro Beltran, Felicia Cao, Bang-Xing Hong, Tania Rodriguez-Cruz, Xiao-Tong Song, Stephen Gottschalk, Hugo Calderon, Sam Illingworth, Alice Brown, Kerry Fisher, Len Seymour, Brian Champion, Emma Eriksson, Jessica Wenthe, Ann-Charlotte Hellström, Gabriella Paul-Wetterberg, Angelica Loskog, Ioanna Milenova, Magnus Ståhle, Justyna Jarblad-Leja, Gustav Ullenhag, Anna Dimberg, Rafael Moreno, Ramon Alemany, Sharad Goyal, Ann Silk, Janice Mehnert, Nashat Gabrail, Jennifer Bryan, Daniel Medina, Leah Mitchell, Kader Yagiz, Fernando Lopez, Daniel Mendoza, Anthony Munday, Harry Gruber, Douglas Jolly, Steven Fuhrmann, Sasa Radoja, Wei Tan, Aldo Pourchet, Alan Frey, Ian Mohr, Matthew Mulvey, Robert H. I. Andtbacka, Merrick Ross, Sanjiv Agarwala, Kenneth Grossmann, Matthew Taylor, John Vetto, Rogerio Neves, Adil Daud, Hung Khong, Stephanie M. Meek, Richard Ungerleider, Scott Welden, Maki Tanaka, Matthew Williams, Sigrun Hallmeyer, Bernard Fox, Zipei Feng, Christopher Paustian, Carlo Bifulco, Sadia Zafar, Otto Hemminki, Simona Bramante, Lotta Vassilev, Hongjie Wang, Andre Lieber, Silvio Hemmi, Tanja de Gruijl, Anna Kanerva, Tameem Ansari, Srividya Sundararaman, Diana Roen, Paul Lehmann, Anja C. Bloom, Lewis H. Bender, Ian B. Walters, Jay A. Berzofsky, Fanny Chapelin, Eric T. Ahrens, Jeff DeFalco, Michael Harbell, Amy Manning-Bog, Alexander Scholz, Danhui Zhang, Gilson Baia, Yann Chong Tan, Jeremy Sokolove, Dongkyoon Kim, Kevin Williamson, Xiaomu Chen, Jillian Colrain, Gregg Espiritu Santo, Ngan Nguyen, Wayne Volkmuth, Norman Greenberg, William Robinson, Daniel Emerling, Charles G. Drake, Daniel P. Petrylak, Emmanuel S. Antonarakis, Adam S. Kibel, Nancy N. Chang, Tuyen Vu, Dwayne Campogan, Heather Haynes, James B. Trager, Nadeem A. Sheikh, David I. Quinn, Peter Kirk, Murali Addepalli, Thomas Chang, Ping Zhang, Marina Konakova, Katsunobu Hagihara, Steven Pai, Laurie VanderVeen, Palakshi Obalapur, Peiwen Kuo, Phi Quach, Lawrence Fong, Deborah H. Charych, Jonathan Zalevsky, John L. Langowski, Yolanda Kirksey, Ravi Nutakki, Shalini Kolarkar, Rhoneil Pena, Ute Hoch, Stephen K. Doberstein, John Cha, Zach Mallon, Myra Perez, Amanda McDaniel, Snjezana Anand, Darrin Uecker, Richard Nuccitelli, Eva Wieckowski, Ravikumar Muthuswamy, Roshni Ravindranathan, Ariana N. Renrick, Menaka Thounaojam, Portia Thomas, Samuel Pellom, Anil Shanker, Duafalia Dudimah, Alan Brooks, Yu-Lin Su, Tomasz Adamus, Qifang Zhang, Sergey Nechaev, Marcin Kortylewski, Spencer Wei, Clark Anderson, Chad Tang, Jonathan Schoenhals, Efrosini Tsouko, John Heymach, Patricia de Groot, Joe Chang, Kenneth R. Hess, Adi Diab, Padmanee Sharma, David Hong, James Welsh, Andrea J. Parsons, Jardin Leleux, Stephane Ascarateil, Marie Eve Koziol, Dina Bai, Peihong Dai, Weiyi Wang, Ning Yang, Stewart Shuman, Liang Deng, Patrick Dillon, Gina Petroni, David Brenin, Kim Bullock, Walter Olson, Mark E. Smolkin, Kelly Smith, Carmel Nail, Craig L. Slingluff, Meenu Sharma, Faisal Fa’ak, Louise Janssen, Hiep Khong, Zhilan Xiao, Yared Hailemichael, Manisha Singh, Christina Vianden, Willem W. Overwijk, Andrea Facciabene, Pierini Stefano, Fang Chongyung, Stavros Rafail, Michael Nielsen, Peter Vanderslice, Darren G. Woodside, Robert V. Market, Ronald J. Biediger, Upendra K. Marathi, Kevin Hollevoet, Nick Geukens, Paul Declerck, Nathalie Joly, Laura McIntosh, Eustache Paramithiotis, Magnus Rizell, Malin Sternby, Bengt Andersson, Alex Karlsson-Parra, Rui Kuai, Lukasz Ochyl, Anna Schwendeman, James Moon, Weiwen Deng, Thomas E. Hudson, Bill Hanson, Chris S. Rae, Joel Burrill, Justin Skoble, George Katibah, Michele deVries, Peter Lauer, Thomas W. Dubensky, Xin Chen, Li Zhou, Xiubao Ren, Charu Aggarwal, Drishty Mangrolia, Roger Cohen, Gregory Weinstein, Matthew Morrow, Joshua Bauml, Kim Kraynyak, Jean Boyer, Jian Yan, Jessica Lee, Laurent Humeau, Sandra Oyola, Susan Duff, David Weiner, Zane Yang, Mark Bagarazzi, Douglas G. McNeel, Jens Eickhoff, Robert Jeraj, Mary Jane Staab, Jane Straus, Brian Rekoske, Glenn Liu, Marit Melssen, William Grosh, Nikole Varhegyi, Nadejda Galeassi, Donna H. Deacon, Elizabeth Gaughan, Maurizio Ghisoli, Minal Barve, Robert Mennel, Gladice Wallraven, Luisa Manning, Neil Senzer, John Nemunaitis, Masahiro Ogasawara, Shuichi Ota, Kaitlin M. Peace, Diane F. Hale, Timothy J. Vreeland, Doreen O. Jackson, John S. Berry, Alfred F. Trappey, Garth S. Herbert, Guy T. Clifton, Mark O. Hardin, Anne Toms, Na Qiao, Jennifer Litton, George E. Peoples, Elizabeth A. Mittendorf, Lila Ghamsari, Emilio Flano, Judy Jacques, Biao Liu, Jonathan Havel, Vladimir Makarov, Timothy A. Chan, Jessica B. Flechtner, John Facciponte, Stefano Ugel, Francesco De Sanctis, George Coukos, Sébastien Paris, Agnes Pottier, Laurent Levy, Bo Lu, Federica Cappuccini, Emily Pollock, Richard Bryant, Freddie Hamdy, Adrian Hill, Irina Redchenko, Hussein Sultan, Takumi Kumai, Valentyna Fesenkova, Esteban Celis, Ingrid Fernando, Claudia Palena, Justin M. David, Elizabeth Gabitzsch, Frank Jones, James L. Gulley, Mireia Uribe Herranz, Hiroshi Wada, Atsushi Shimizu, Toshihiro Osada, Satoshi Fukaya, Eiji Sasaki, Milad Abolhalaj, David Askmyr, Kristina Lundberg, Ann-Sofie Albrekt, Lennart Greiff, Malin Lindstedt, Dallas B. Flies, Tomoe Higuchi, Wojciech Ornatowski, Jaryse Harris, Sarah F. Adams, Todd Aguilera, Marjan Rafat, Laura Castellini, Hussein Shehade, Mihalis Kariolis, Dadi Jang, Rie vonEbyen, Edward Graves, Lesley Ellies, Erinn Rankin, Albert Koong, Amato Giaccia, Reham Ajina, Shangzi Wang, Jill Smith, Mariaelena Pierobon, Sandra Jablonski, Emanuel Petricoin, Louis M. Weiner, Lorcan Sherry, John Waller, Mark Anderson, Alison Bigley, Chantale Bernatchez, Cara Haymaker, Harriet Kluger, Michael Tetzlaff, Natalie Jackson, Ivan Gergel, Mary Tagliaferri, Patrick Hwu, Mario Snzol, Michael Hurwitz, Theresa Barberi, Allison Martin, Rahul Suresh, David Barakat, Sarah Harris-Bookman, Charles Drake, Alan Friedman, Sara Berkey, Stephanie Downs-Canner, Robert P. Edwards, Tyler Curiel, Kunle Odunsi, Tullia C. Bruno, Brandon Moore, Olivia Squalls, Peggy Ebner, Katherine Waugh, John Mitchell, Wilbur Franklin, Daniel Merrick, Martin McCarter, Brent Palmer, Jeffrey Kern, Dario Vignali, Jill Slansky, Anissa S. H. Chan, Xiaohong Qiu, Kathryn Fraser, Adria Jonas, Nadine Ottoson, Keith Gordon, Takashi O. Kangas, Steven Leonardo, Kathleen Ertelt, Richard Walsh, Mark Uhlik, Jeremy Graff, Nandita Bose, Ravi Gupta, Nitin Mandloi, Kiran Paul, Ashwini Patil, Rekha Sathian, Aparna Mohan, Malini Manoharan, Amitabha Chaudhuri, Yu Chen, Jing Lin, Yun-bin Ye, Chun-wei Xu, Gang Chen, Zeng-qing Guo, Andrey Komarov, Alex Chenchik, Michael Makhanov, Costa Frangou, Yi Zheng, Carla Coltharp, Darryn Unfricht, Ryan Dilworth, Leticia Fridman, Linying Liu, Milind Rajopadhye, Peter Miller, Fernando Concha-Benavente, Julie Bauman, Sumita Trivedi, Raghvendra Srivastava, James Ohr, Dwight Heron, Uma Duvvuri, Seungwon Kim, Heather Torrey, Toshi Mera, Yoshiaki Okubo, Eva Vanamee, Rosemary Foster, Denise Faustman, Edward Stack, Daisuke Izaki, Kristen Beck, Dan Tong Jia, Paul Armenta, Ashley White-Stern, Douglas Marks, Bret Taback, Basil Horst, Laura Hix Glickman, David B. Kanne, Kelsey S. Gauthier, Anthony L. Desbien, Brian Francica, Justin L. Leong, Leonard Sung, Ken Metchette, Shailaja Kasibhatla, Anne Marie Pferdekamper, Lianxing Zheng, Charles Cho, Yan Feng, Jeffery M. McKenna, John Tallarico, Steven Bender, Chudi Ndubaku, Sarah M. McWhirter, Elena Gonzalez Gugel, Charles J. M. Bell, Adiel Munk, Luciana Muniz, Nina Bhardwaj, Fei Zhao, Kathy Evans, Christine Xiao, Alisha Holtzhausen, Brent A. Hanks, Nathalie Scholler, Catherine Yin, Pien Van der Meijs, Andrew M. Prantner, Cecile M. Krejsa, Leia Smith, Brian Johnson, Daniel Branstetter, Paul L. Stein, Juan C. Jaen, Joanne BL Tan, Ada Chen, Timothy Park, Jay P. Powers, Holly Sexton, Guifen Xu, Steve W. Young, Ulrike Schindler, Wentao Deng, David John Klinke, Hannah M. Komar, Gregory Serpa, Omar Elnaggar, Philip Hart, Carl Schmidt, Mary Dillhoff, Ming Jin, Michael C. Ostrowski, Madhuri Koti, Katrina Au, Nichole Peterson, Peter Truesdell, Gillian Reid-Schachter, Charles Graham, Andrew Craig, Julie-Ann Francis, Beatrix Kotlan, Timea Balatoni, Emil Farkas, Laszlo Toth, Mihaly Ujhelyi, Akos Savolt, Zoltan Doleschall, Szabolcs Horvath, Klara Eles, Judit Olasz, Orsolya Csuka, Miklos Kasler, Gabriella Liszkay, Eytan Barnea, Collin Blakely, Patrick Flynn, Reid Goodman, Raphael Bueno, David Sugarbaker, David Jablons, V. Courtney Broaddus, Brian West, Paul R. Kunk, Joseph M. Obeid, Kevin Winters, Patcharin Pramoonjago, Edward B. Stelow, Todd W. Bauer, Osama E. Rahma, Adam Lamble, Yoko Kosaka, Fei Huang, Kate A. Saser, Homer Adams, Christina E. Tognon, Ted Laderas, Shannon McWeeney, Marc Loriaux, Jeffery W. Tyner, Brian J. Druker, Evan F. Lind, Zhuqing Liu, Shanhong Lu, Lawrence P. Kane, Gulidanna Shayan, Julia Femel, Ryan Lane, Jamie Booth, Amanda W. Lund, Anthony Rodriguez, Victor H. Engelhard, Alessandra Metelli, Bill X. Wu, Caroline W. Fugle, Rachidi Saleh, Shaoli Sun, Jennifer Wu, Bei Liu, Zihai Li, Zachary S. Morris, Emily I. Guy, Clinton Heinze, Jasdeep Kler, Monica M. Gressett, Lauryn R. Werner, Stephen D. Gillies, Alan J. Korman, Hans Loibner, Jacquelyn A. Hank, Alexander L. Rakhmilevich, Paul M. Harari, Paul M. Sondel, Erica Huelsmann, Joseph Broucek, Dorothee Brech, Tobias Straub, Martin Irmler, Johannes Beckers, Florian Buettner, Elke Schaeffeler, Matthias Schwab, Elfriede Noessner, Alison Wolfreys, Andre Da Costa, John Silva, Andrea Crosby, Ludovicus Staelens, Graham Craggs, Annick Cauvin, Sean Mason, Alison M. Paterson, Andrew C. Lake, Caroline M. Armet, Rachel W. O’Connor, Jonathan A. Hill, Emmanuel Normant, Ammar Adam, Detlev M. Biniszkiewicz, Scott C. Chappel, Vito J. Palombella, Pamela M. Holland, Annette Becker, Manmohan R. Leleti, Eric Newcomb, Joanne B. L. Tan, Suthee Rapisuwon, Arash Radfar, Kellie Gardner, Geoffrey Gibney, Michael Atkins, Keith R. Rennier, Robert Crowder, Ping Wang, Russell K. Pachynski, Rosa M. Santana Carrero, Sarai Rivas, Figen Beceren-Braun, Scott Anthony, Kimberly S. Schluns, Deepali Sawant, Maria Chikina, Hiroshi Yano, Creg Workman, Elise Salerno, Ileana Mauldin, Donna Deacon, Sofia Shea, Joel Pinczewski, Thomas Gajewski, Stefani Spranger, Brendan Horton, Akiko Suzuki, Pamela Leland, Bharat H. Joshi, Raj K. Puri, Randy F. Sweis, Riyue Bao, Jason Luke, Marie-Nicole Theodoraki, Frances-Mary Mogundo, Haejung Won, Dayson Moreira, Chan Gao, Xingli Zhao, Priyanka Duttagupta, Jeremy Jones, Massimo D’Apuzzo, and Sumanta Pal
- Subjects
0301 basic medicine ,Pharmacology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Cancer ,Immunotherapy ,medicine.disease ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business - Abstract
O1 IL-15 primes an mTOR-regulated gene-expression program to prolong anti-tumor capacity of human natural killer cells #### Andreas Lundqvist1, Vincent van Hoef1, Xiaonan Zhang1, Erik Wennerberg2, Julie Lorent1, Kristina Witt1, Laia Masvidal Sanz1, Shuo Liang1, Shannon Murray3, Ola Larsson1
- Published
- 2016
37. Molecular Imaging of Immunotherapy Targets in Cancer
- Author
-
Christopher G. England, Weibo Cai, Douglas G. McNeel, and Emily B. Ehlerding
- Subjects
0301 basic medicine ,Noninvasive imaging ,medicine.medical_treatment ,Focus on Molecular Imaging ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,PD-L1 ,Neoplasms ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Molecular Targeted Therapy ,biology ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,Molecular Imaging ,030104 developmental biology ,CTLA-4 ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,biology.protein ,Molecular imaging ,business ,Preclinical imaging - Abstract
Immunotherapy has emerged as a promising alternative in the arsenal against cancer by harnessing the power of the immune system to specifically target malignant tissues. As the field of immunotherapy continues to expand, researchers will require newer methods for studying the interactions between the immune system, tumor cells, and immunotherapy agents. Recently, several noninvasive imaging strategies have been used to map the biodistribution of immune checkpoint molecules, monitor the efficacy and potential toxicities of the treatments, and identify patients who are likely to benefit from immunotherapies. In this review, we outline the current applications of noninvasive techniques for the preclinical imaging of immunotherapy targets and suggest future pathways for molecular imaging to contribute to this developing field.
- Published
- 2016
38. Antitumor vaccination of prostate cancer patients elicits PD-1/PD-L1 regulated antigen-specific immune responses
- Author
-
Douglas G. McNeel, Brian M. Olson, and Brian T. Rekoske
- Subjects
0301 basic medicine ,Receptor expression ,Immunology ,Biology ,medicine.disease ,DNA vaccination ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Circulating tumor cell ,Oncology ,Immunization ,Antigen ,030220 oncology & carcinogenesis ,Blocking antibody ,Cancer research ,medicine ,Immunology and Allergy ,Original Research - Abstract
We have previously reported that tumor antigen-specific DNA vaccination in mice led to an increase in IFNγ-secreting T cells and an increase in tumor expression of PD-L1. Further, we demonstrated that increasing the encoded antigen's MHC-binding affinity led to increased PD-1 expression on antigen-specific CD8(+) T cells. Together these phenomena provided resistance to antitumor immunization that was abrogated with PD-1/PD-L1 blockade. We consequently sought to determine whether similar regulation occurred in human patients following antitumor immunization. Using clinical samples from prostate cancer patients who were previously immunized with a DNA vaccine, we analyzed changes in checkpoint receptor expression on antigen-specific CD8(+) T cells, the effect of PD-1 blockade on elicited immune responses, and for changes in checkpoint ligand expression on patients' circulating tumor cells (CTCs). We observed no significant changes in T-cell expression of PD-1 or other checkpoint receptors, but antigen-specific immune responses were detected and/or augmented with PD-1 blockade as detected by IFNγ and granzyme B secretion or trans vivo DTH testing. Moreover, PD-L1 expression was increased on CTCs following vaccination, and this PD-L1 upregulation was associated with the development of sustained T-cell immunity and longer progression-free survival. Finally, similar results were observed with patients treated with sipuleucel-T, another vaccine targeting the same prostate antigen. These findings provide in-human rationale for combining anticancer vaccines with PD-1 blocking antibodies, particularly for the treatment of prostate cancer, a disease for which vaccines have demonstrated benefit and yet PD-1 inhibitors have shown little clinical benefit to date as monotherapies.
- Published
- 2016
39. Phase I trial of tremelimumab in combination with short-term androgen deprivation in patients with PSA-recurrent prostate cancer
- Author
-
Jens C. Eickhoff, Heath A. Smith, Joshua M. Lang, George Wilding, Glenn Liu, Douglas G. McNeel, and Mary Jane Staab
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Bicalutamide ,medicine.drug_class ,Immunology ,Antibodies, Monoclonal, Humanized ,urologic and male genital diseases ,Antiandrogen ,Article ,Tosyl Compounds ,Androgen deprivation therapy ,Prostate cancer ,Prostate ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Nitriles ,medicine ,Humans ,Immunology and Allergy ,Anilides ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Antibodies, Monoclonal ,Prostatic Neoplasms ,Androgen Antagonists ,Middle Aged ,Prostate-Specific Antigen ,medicine.disease ,Androgen ,Prostate-specific antigen ,Treatment Outcome ,medicine.anatomical_structure ,Disease Progression ,business ,Tremelimumab ,medicine.drug - Abstract
CTLA-4 blockade has demonstrated antitumor efficacy in human clinical trials. The antitumor mechanism is presumably mediated in part by the expansion of tumor-specific T cells. Androgen deprivation, the cornerstone of treatment for patients with metastatic prostate cancer, has been shown to elicit prostate tissue apoptosis and lymphocytic inflammation. We hypothesized that treatment with androgen deprivation, followed by an anti-CTLA-4 antibody, could augment a tumor-specific immune response elicited by androgen deprivation. We report here the results of a phase I trial evaluating a humanized monoclonal antibody targeting CTLA-4, CP-675,206 (tremelimumab), in combination with androgen deprivation using an antiandrogen. Eligible patients were those with PSA-recurrent prostate cancer after primary surgery and/or radiation therapy, not previously treated with androgen deprivation, and without radiographic evidence of metastatic disease. Subjects were treated in two cycles, 3 months apart, in which they received bicalutamide 150 mg daily days 1–28 and tremelimumab on day 29. The primary endpoint of the trial was safety. Secondary endpoints included measures of PSA kinetics and identification of a maximum tolerated dose. Eleven patients were enrolled and completed at least 1 year of follow-up. Dose-limiting toxicities included grade 3 diarrhea and skin rash. No favorable changes in PSA doubling time were observed in a period shortly after completing treatment; however, three patients experienced a prolongation in PSA doubling time detectable several months after completing treatment. The identification of delayed, prolonged favorable changes in serum PSA suggests that future studies could explore this combination in studies evaluating time to disease progression.
- Published
- 2011
40. Vaccines Targeting the Cancer-testis Antigen SSX-2 Elicit HLA-A2 Epitope-specific Cytolytic T Cells
- Author
-
Heath A. Smith and Douglas G. McNeel
- Subjects
Male ,Cancer Research ,medicine.medical_treatment ,Immunology ,Mice, Transgenic ,Human leukocyte antigen ,Immunodominance ,Biology ,Cancer Vaccines ,Article ,Epitope ,Epitopes ,Mice ,Prostate cancer ,Antigen ,Cell Line, Tumor ,HLA-A2 Antigen ,Vaccines, DNA ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Amino Acid Sequence ,Cells, Cultured ,Pharmacology ,HLA-DR1 Antigen ,Prostatic Neoplasms ,Immunotherapy ,medicine.disease ,Neoplasm Proteins ,Mice, Inbred C57BL ,Repressor Proteins ,Leukocytes, Mononuclear ,Cancer/testis antigens ,Peptides ,T-Lymphocytes, Cytotoxic - Abstract
The cancer-testis antigen synovial sarcoma X breakpoint-2 (SSX-2) is a potentially attractive target for tumor immunotherapy based upon its tissue-restricted expression to germline cells and its frequent expression in malignancies. The goal of this study was to evaluate genetic vaccine encoding SSX-2 to prioritize human leukocyte antigen (HLA)-A2-specific epitopes and determine if a DNA vaccine can elicit SSX-2-specific cytotoxic T lymphocytes (CTLs) capable of lysing prostate cancer cells. HLA-A2-restricted epitopes were identified based on their in vitro binding affinity for HLA-A2 and by the ability of a genetic vaccine to elicit peptide-specific CTL in A2/DR1 (HLA-A2.1+/HLA-DR1+/H-2 class I-/class II-knockout) transgenic mice. We found that SSX-2 peptides p41-49 (KASEKIFYV) and p103-111 (RLQGISPKI) had high affinity for HLA-A2 and were immunogenic in vivo; however, peptide p103-111 was immunodominant with robust peptide-specific immune responses elicited in mice vaccinated with a plasmid DNA vaccine encoding SSX-2. Furthermore, p103-111-specific CTLs were able to lyse an HLA-A2+ prostate cancer cell line. The immunodominance of this epitope was found not to be due to a putative HLA-DR1 epitope (p98-112) flanking p103-111. Finally, we demonstrated that SSX-2 epitope-specific CTLs could be detected and cultured from the peripheral blood of HLA-A2+ prostate cancer patients, notably patients with advanced prostate cancer. Overall, we conclude that SSX-2 peptide p103-111 is an immunodominant HLA-A2-restricted epitope, and epitope-specific CD8 T cells can be detected in patients with prostate cancer, suggesting that tolerance to SSX-2 can be circumvented in vivo. Together, these findings suggest that SSX-2 may be a relevant target antigen for prostate cancer vaccine approaches.
- Published
- 2011
41. Identification of prostatic acid phosphatase (PAP) specific HLA-DR1-restricted t-cell epitopes
- Author
-
Douglas G. McNeel and Laura E. Johnson
- Subjects
business.industry ,Urology ,ELISPOT ,Active immunotherapy ,medicine.disease ,Epitope ,Tumor antigen ,DNA vaccination ,Prostate cancer ,Epitope mapping ,Oncology ,Prostatic acid phosphatase ,Immunology ,Medicine ,business - Abstract
BACKGROUND Prostatic acid phosphatase (PAP) is a prostate cancer tumor antigen and is an immunological target in several active immunotherapy clinical trials for the treatment of prostate cancer. We and others have demonstrated that PAP-specific T-cell responses can be elicited and augmented following antigen-specific immunization in both humans and animal models. We have previously reported that prostate cancer patients immunized with a DNA vaccine encoding PAP (pTVG-HP) developed both CD4+ and CD8+ T-cell responses. PAP-specific, CD4+ T-cell proliferative responses were generated in three out of four HLA-DRB1*0101 patients suggesting the possibility that DR1-restricted epitopes exist. METHODS To identify PAP-specific HLA-DRB1*0101 restricted epitopes, we immunized HLA-A2.01/HLA-DRB1*0101 (A2/DR1) transgenic mice with the pTVG-HP DNA vaccine. To map DRB1*0101-restricted epitopes, splenocytes from immunized mice were screened against a library of overlapping 15-residue, PAP-derived peptides using an IFNγ ELISPOT assay. RESULTS We identified four HLA-DRB1*0101 epitopes for PAP in A2/DR1 mice (PAP161–175, PAP181–195, PAP191–205, and PAP 351–365). T cells specific for one epitope (PAP181–195) were found to be augmented after immunization in a HLA-DRB1*0101+ prostate cancer patient. CONCLUSIONS The identification of MHC class II epitopes may provide tools to directly monitor immune responses after vaccination and may be important for the design of future prostate cancer vaccines. Prostate 72:730–740, 2012. © 2011 Wiley Periodicals, Inc.
- Published
- 2011
42. Lenalidomide modulates IL-8 and anti-prostate antibody levels in men with biochemically recurrent prostate cancer
- Author
-
Heath A. Smith, Marianna Zahurak, Daniel Keizman, Emmanuel S. Antonarakis, Mario A. Eisenberger, Michael A. Carducci, Douglas G. McNeel, Christopher J. Thoburn, Charles G. Drake, and Daniel J. Zabransky
- Subjects
business.industry ,Prostatectomy ,Urology ,medicine.medical_treatment ,medicine.disease ,Thalidomide ,Prostate cancer ,Prostate-specific antigen ,medicine.anatomical_structure ,Cytokine ,Oncology ,Prostate ,Immunology ,Medicine ,Adenocarcinoma ,business ,Lenalidomide ,medicine.drug - Abstract
BACKGROUND We retrospectively explored changes in immunological parameters in men with biochemically recurrent prostate cancer treated with either 5 or 25 mg of lenalidomide in a randomized phase 2 trial, and determined whether those changes correlated with disease progression. METHODS Cytokine levels were compared for each patient at baseline and after 6 months of treatment with lenalidomide. Regression models for correlated data were used to assess associations of cytokine levels with lenalidomide treatment effect. Estimates were obtained using generalized estimating equations. Changes in circulating anti-prostate antibodies were evaluated using a high-throughput immunoblot technique. RESULTS Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the TH2 cytokines IL-4, IL-5, IL-10, and IL-13. Disease progression in treated patients was associated with an increase in circulating IL-8 levels, while IL-8 levels decreased significantly in non-progressors. CONCLUSIONS Lenalidomide demonstrates immunomodulatory properties in patients with biochemically recurrent prostate cancer. The induction of novel anti-prostate antibodies is a potential mechanism for lenalidomide response. Changes in serum IL-8 levels may serve as a potential biomarker in treated patients. These hypotheses require formal testing in future prospective trials. Prostate 72:487–498, 2012. © 2011 Wiley Periodicals, Inc.
- Published
- 2011
43. CD8+ T cells specific for the androgen receptor are common in patients with prostate cancer and are able to lyse prostate tumor cells
- Author
-
Brian M. Olson and Douglas G. McNeel
- Subjects
Male ,Cancer Research ,medicine.medical_treatment ,Immunology ,Epitopes, T-Lymphocyte ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Biology ,Ligands ,Article ,Epitope ,Animals, Genetically Modified ,Mice ,Prostate cancer ,Antigen ,Cell Line, Tumor ,HLA-A2 Antigen ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Amino Acid Sequence ,Mice, Knockout ,HLA-DR1 Antigen ,Prostatic Neoplasms ,Cancer ,Immunotherapy ,medicine.disease ,Protein Structure, Tertiary ,CTL ,Oncology ,Receptors, Androgen ,Cytokines ,Oligopeptides ,CD8 ,T-Lymphocytes, Cytotoxic - Abstract
The androgen receptor (AR) is a hormone receptor that plays a critical role in prostate cancer, and depletion of its ligand has long been the cornerstone of treatment for metastatic disease. Here, we evaluate the AR ligand-binding domain (LBD) as an immunological target, seeking to identify HLA-A2-restricted epitopes recognized by T cells in prostate cancer patients. Ten AR LBD-derived, HLA-A2-binding peptides were identified and ranked with respect to HLA-A2 affinity and were used to culture peptide-specific T cells from HLA-A2+ prostate cancer patients. These T-cell cultures identified peptide-specific T cells specific for all ten peptides in at least one patient, and T cells specific for peptides AR805 and AR811 were detected in over half of patients. Peptide-specific CD8+ T-cell clones were then isolated and characterized for prostate cancer cytotoxicity and cytokine expression, identifying that AR805 and AR811 CD8+ T-cell clones could lyse prostate cancer cells in an HLA-A2-restricted fashion, but only AR811 CTL had polyfunctional cytokine expression. Epitopes were confirmed using immunization studies in HLA-A2 transgenic mice, in which the AR LBD is an autologous antigen with an identical protein sequence, which showed that mice immunized with AR811 developed peptide-specific CTL that lyse HLA-A2+ prostate cancer cells. These data show that AR805 and AR811 are HLA-A2-restricted epitopes for which CTL can be commonly detected in prostate cancer patients. Moreover, CTL responses specific for AR811 can be elicited by direct immunization of A2/DR1 mice. These findings suggest that it may be possible to elicit an anti-prostate tumor immune response by augmenting CTL populations using AR LBD-based vaccines.
- Published
- 2011
44. Antibody responses to prostate-associated antigens in patients with prostatitis and prostate cancer
- Author
-
Brett B. Maricque, Douglas G. McNeel, and Jens C. Eickhoff
- Subjects
PCA3 ,biology ,business.industry ,Urology ,medicine.medical_treatment ,Cancer ,Prostatitis ,Immunotherapy ,medicine.disease ,Immunoglobulin G ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,Antigen ,Prostate ,Immunology ,medicine ,biology.protein ,business - Abstract
BACKGROUND An important focus of tumor immunotherapy has been the identification of appropriate antigenic targets. Serum-based screening approaches have led to the discovery of hundreds of tumor-associated antigens recognized by IgG. Our efforts to identify immunologically recognized proteins in prostate cancer have yielded a multitude of antigens; however, prioritizing these antigens as targets for evaluation in immunotherapies has been challenging. In this report, we set out to determine whether the evaluation of multiple antigenic targets would allow the identification of a subset of antigens that are common immunologic targets in patients with prostate cancer. METHODS Using a phage immunoblot approach, we evaluated IgG responses in patients with prostate cancer (n = 126), patients with chronic prostatitis (n = 45), and men without prostate disease (n = 53). RESULTS We found that patients with prostate cancer or prostatitis have IgG specific for multiple common antigens. A subset of 23 proteins was identified to which IgG were detected in 38% of patients with prostate cancer and 33% patients with prostatitis versus 6% of controls (P
- Published
- 2010
45. DNA vaccines for the treatment of prostate cancer
- Author
-
Douglas G. McNeel and Sheeba Alam
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Immunology ,Context (language use) ,Disease ,Cancer Vaccines ,DNA vaccination ,Prostate cancer ,Antigen ,Serum biomarkers ,Internal medicine ,Drug Discovery ,Vaccines, DNA ,medicine ,Animals ,Humans ,Pharmacology ,Clinical Trials as Topic ,business.industry ,Prostatic Neoplasms ,Cancer ,medicine.disease ,Clinical trial ,Disease Models, Animal ,Molecular Medicine ,Immunotherapy ,business - Abstract
Prostate cancer is a significant public health problem, and the most commonly diagnosed cancer in the USA. The long natural history of prostate cancer, the presence of a serum biomarker that can be used to detect very early recurrences, and the previous identification of multiple potential tissue-specific target antigens are all features that make this disease suitable for the development of anti-tumor vaccines. To date, many anti-tumor vaccines have entered clinical testing for patients with prostate cancer, and some have demonstrated clinical benefit. DNA vaccines represent one vaccine approach that has been evaluated in multiple preclinical models and clinical trials. The safety, specificity for the target antigen, ease of manufacturing and ease of incorporating other immune-modulating approaches make DNA vaccines particularly relevant for future development. This article focuses on DNA vaccines specifically in the context of prostate cancer treatment, focusing on antigens targeted in preclinical models, recent clinical trials and efforts to improve the potency of these vaccines.
- Published
- 2010
46. Heterologous vaccination targeting prostatic acid phosphatase (PAP) using DNA and Listeria vaccines elicits superior anti-tumor immunity dependent on CD4+ T cells elicited by DNA priming
- Author
-
John-Demian Sauer, Dirk G. Brockstedt, Douglas G. McNeel, Meredith L. Leong, Erin Theisen, Peter Lauer, and Laura E. Johnson
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Cellular immunity ,listeria monocytogenes ,Immunology ,Priming (immunology) ,Heterologous ,heterologous prime-boost ,lcsh:RC254-282 ,DNA vaccination ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Immunity ,Immunology and Allergy ,Medicine ,Original Research ,business.industry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Vaccination ,030104 developmental biology ,Oncology ,dna vaccine ,lcsh:RC581-607 ,business ,prostatic acid phosphatase ,030215 immunology - Abstract
Background. Sipuleucel T, an autologous cell-based vaccine targeting prostatic acid phosphatase (PAP), has demonstrated efficacy for the treatment of advanced prostate cancer. DNA vaccines encoding PAP and live attenuated Listeria vaccines have entered clinical trials for patients with prostate cancer, and have advantages in terms of eliciting predominantly Th1-biased immunity. In this study, we investigated whether the immunogenicity and anti-tumor efficacy of a DNA and Listeria vaccine, each encoding PAP, could be enhanced by using them in a heterologous prime/boost approach. Methods. Transgenic mice expressing HLA-A2.01 and HLA-DRB1*0101 were immunized alone or with a heterologous prime/boost strategy. Splenocytes were evaluated for MHC class I and II-restricted, PAP-specific immune responses by IFNγ ELISPOTs. Anti-tumor activity to a syngeneic, PAP-expressing tumor line was evaluated. Results. PAP-specific cellular immunity and anti-tumor activity were elicited in mice after immunization with DNA- or listeria-based vaccines. Greater CD4+ and CD8+ responses, and anti-tumor responses, were elicited when mice were immunized first with DNA and boosted with Listeria, but not when administered in the opposite order. This was found to be dependent on CD4+ T cells elicited with DNA priming, and was not due to inflammatory signals by Listeria itself or due to B cells serving as antigen-presenting cells for DNA during priming. Conclusions. Heterologous prime/boost vaccination using DNA priming with Listeria boosting may provide better anti-tumor immunity, similar to many reports evaluating DNA priming with vaccines targeting foreign microbial antigens. These findings have implications for the design of future clinical trials.
- Published
- 2018
47. HLA-A2-restricted T-cell epitopes specific for prostatic acid phosphatase
- Author
-
Laura E. Johnson, Mary L. Disis, Thomas Frye, Brian M. Olson, Douglas G. McNeel, Lawrence Fong, and Keith L. Knutson
- Subjects
Male ,Cancer Research ,Acid Phosphatase ,Immunology ,Epitopes, T-Lymphocyte ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Cancer Vaccines ,Article ,Epitope ,Interferon-gamma ,Mice ,Immune system ,Antigen ,Antigens, Neoplasm ,HLA-A2 Antigen ,MHC class I ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,ELISPOT ,Prostatic Neoplasms ,Virology ,Peptide Fragments ,Clone Cells ,Oncology ,Prostatic acid phosphatase ,biology.protein ,Immunization ,Protein Tyrosine Phosphatases ,CD8 ,Protein Binding - Abstract
Prostatic acid phosphatase (PAP) has been investigated as the target of several antigen-specific anti-prostate tumor vaccines. The goal of antigen-specific active immunotherapies targeting PAP would ideally be to elicit PAP-specific CD8+ effector T cells. The identification of PAP-specific CD8+ T-cell epitopes should provide a means of monitoring the immunological efficacy of vaccines targeting PAP, and these epitopes might themselves be developed as vaccine antigens. In the current report, we hypothesized that PAP-specific epitopes might be identified by direct identification of pre-existing CD8+ T cells specific for HLA-A2-restricted peptides derived from PAP in the blood of HLA-A2-expressing individuals. 11 nonamer peptides derived from the amino acid sequence of PAP were used as stimulator antigens in functional ELISPOT assays with peripheral blood mononuclear cells from 20 HLA-A2+ patients with prostate cancer or ten healthy blood donors. Peptide-specific T cells were frequently identified in both groups for three of the peptides, p18-26, p112-120, and p135-143. CD8+ T-cell clones specific for three peptides, p18-26, p112-120, and p299-307, confirmed that these are HLA-A2-restricted T-cell epitopes. Moreover, HLA-A2 transgenic mice immunized with a DNA vaccine encoding PAP developed epitope-specific responses for one or more of these three peptide epitopes. We propose that this method to first identify epitopes for which there are pre-existing epitope-specific T cells could be used to prioritize MHC class I-specific epitopes for other antigens. In addition, we propose that the epitopes identified here could be used to monitor immune responses in HLA-A2+ patients receiving vaccines targeting PAP to identify potentially therapeutic immune responses.
- Published
- 2010
48. The SSX Family of Cancer-Testis Antigens as Target Proteins for Tumor Therapy
- Author
-
Douglas G. McNeel and Heath A. Smith
- Subjects
lcsh:Immunologic diseases. Allergy ,Cytotoxicity, Immunologic ,Male ,Immunology ,Review Article ,CD8-Positive T-Lymphocytes ,Biology ,Cancer Vaccines ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Antigens, Neoplasm ,Neoplasms ,MHC class I ,medicine ,Humans ,Immunology and Allergy ,030304 developmental biology ,0303 health sciences ,Cancer ,General Medicine ,Prognosis ,medicine.disease ,Synovial sarcoma ,Neoplasm Proteins ,3. Good health ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,030220 oncology & carcinogenesis ,biology.protein ,Cancer/testis antigens ,Female ,Ectopic expression ,Immunotherapy ,lcsh:RC581-607 ,CD8 - Abstract
Cancer-testis antigens (CTAs) represent an expanding class of tumor-associated proteins defined on the basis of their tissue-restricted expression to testis or ovary germline cells and frequent ectopic expression in tumor tissue. The expression of CTA in MHC class I-deficient germline cells makes these proteins particularly attractive as immunotherapeutic targets because they serve as essentially tumor-specific antigens for MHC class I-restricted CD8+ T cells. Moreover, because CTAs are expressed in many types of cancer, any therapeutic developed to target these antigens might have efficacy for multiple cancer types. Of particular interest among CTAs is the synovial sarcoma X chromosome breakpoint (SSX) family of proteins, which includes ten highly homologous family members. Expression of SSX proteins in tumor tissues has been associated with advanced stages of disease and worse patient prognosis. Additionally, both humoral and cell-mediated immune responses to SSX proteins have been demonstrated in patients with tumors of varying histological origin, which indicates that natural immune responses can be spontaneously generated to these antigens in cancer patients. The current review will describe the history and identification of this family of proteins, as well as what is known of their function, expression in normal and malignant tissues, and immunogenicity.
- Published
- 2010
49. Potentiating Endogenous Antitumor Immunity to Prostate Cancer through Combination Immunotherapy with CTLA4 Blockade and GM-CSF
- Author
-
Shaun O'Brien, Brian D. Kavanagh, Douglas G. McNeel, Eric J. Small, Vivian Weinberg, Brian I. Rini, Amy M. Lin, Lawrence Fong, Jonathan E. Rosenberg, Serena S. Kwek, and Charles J. Ryan
- Subjects
Male ,Cancer Research ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Ipilimumab ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Antigen ,Antigens, CD ,Antigens, Neoplasm ,Humans ,Medicine ,Cytotoxic T cell ,CTLA-4 Antigen ,IL-2 receptor ,Aged ,Aged, 80 and over ,biology ,business.industry ,Antibodies, Monoclonal ,Granulocyte-Macrophage Colony-Stimulating Factor ,Prostatic Neoplasms ,Immunotherapy ,Middle Aged ,Blockade ,Oncology ,Immunology ,Cancer research ,biology.protein ,Antibody ,business ,CD8 ,medicine.drug - Abstract
CTL-associated antigen 4 (CTLA4) is a costimulatory molecule expressed on activated T cells that delivers an inhibitory signal to these T cells. CTLA4 blockade with antibody treatment has been shown to augment antitumor immunity in animal models and is being developed as a treatment for cancer patients. As has been seen in preclinical models, combining CTLA4 blockade and granulocyte macrophage colony-stimulating factor (GM-CSF)–based immunotherapies can enhance the antitumor efficacy of this approach. We therefore examined whether CTLA4 blockade could be combined with GM-CSF administration. We treated 24 patients with metastatic, castration-resistant prostate cancer in a phase I trial where sequential cohorts were treated with increasing doses of ipilimumab, a fully human anti-CTLA4 antibody. Study subjects also received s.c. injections of GM-CSF at a fixed dose. Of the six patients treated at the highest dose level, three had confirmed PSA declines of >50%, including one patient that had a partial response in visceral metastases. Expansion of activated, circulating CD25+ CD69+ CD8+ T cells occurred more frequently at higher doses of treatment and was greater in magnitude than was seen in patients who received the same doses of either ipilimumab or GM-CSF alone. By screening sera with protein arrays, we showed that our treatment can induce antibody responses to NY-ESO-1. These results show that this combination immunotherapy can induce the expansion not only of activated effector CD8 T cells in vivo but also of T cells that are specific for known tumor-associated antigens from the endogenous immune repertoire. [Cancer Res 2009;69(2):609–15]
- Published
- 2009
50. GVAX: an allogeneic, whole-cell, GM-CSF-secreting cellular immunotherapy for the treatment of prostate cancer
- Author
-
Douglas G. McNeel and James E Ward
- Subjects
Male ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Clinical Biochemistry ,Immunotherapy, Adoptive ,Prostate cancer ,Antigens, CD ,Internal medicine ,Drug Discovery ,medicine ,Humans ,CTLA-4 Antigen ,In patient ,Pharmacology ,Clinical Trials as Topic ,business.industry ,Granulocyte-Macrophage Colony-Stimulating Factor ,Prostatic Neoplasms ,Immunotherapy ,medicine.disease ,Antigens, Differentiation ,GVAX ,Clinical trial ,Immunology ,Cellular immunotherapy ,Whole cell ,business ,Clinical evaluation ,Immunosuppressive Agents - Abstract
Antitumor vaccines, also known as active immunotherapies, have become an intense focus of research over the last decade as a means to ideally provide new targeted, and less toxic, therapies to patients with a variety of malignancies. Prostate cancer is among the diseases where a large amount of promising data has been accumulating regarding several distinct active immunotherapies. Here the authors review one of the therapies, known as GVAX, which is among the furthest along in clinical evaluation. The authors review the preclinical studies leading to the development of this immunotherapy, describe the results from clinical trials conducted in patients with prostate cancer, and discuss the benefits, limitations and potential future applications of this technology.
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.