Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
J Immunother Cancer ; 11(11)2023 11 29.
Article in English | MEDLINE | ID: mdl-38030302

ABSTRACT

BACKGROUND: The survival of patients with cervical cancer who are treated with cisplatin in conjunction with the topoisomerase I inhibitor topotecan is enhanced when compared with patients treated with only one of these chemotherapeutics. Moreover, cisplatin-based and T cell-based immunotherapy have been shown to synergize, resulting in stronger antitumor responses. Here, we interrogated whether topotecan could further enhance the synergy of cisplatin with T cell-based cancer immunotherapy. METHODS: Mice bearing human papilloma virus 16 (HPV16) E6/E7-expressing TC-1 tumors were vaccinated with HPV16 E7 long peptides and additionally received chemotherapy consisting of cisplatin and topotecan. We performed an in-depth study of this combinatorial chemoimmunotherapy on the effector function and expansion/contraction kinetics of vaccine-induced CD8+ T cells in the peripheral blood and tumor microenvironment (TME). In addition, we interrogated the particular role of chemotherapy-induced upregulation of costimulatory ligands by tumor-infiltrated myeloid cells on T cell proliferation and survival. RESULTS: We show that E7 long peptide vaccination combined with cisplatin and topotecan, results in CD8+ T cell-dependent durable rejection of established tumors and 94% long-term survival. Although topotecan initially repressed the expansion of vaccine-induced CD8+ T cells, these cells eventually expanded vigorously, which was followed by delayed contraction. These effects associated with the induction of the proliferation marker Ki-67 and the antiapoptosis molecule Bcl-2 by intratumoral tumor-specific CD8+ T cells, which was regulated by topotecan-mediated upregulation of the costimulatory ligand CD70 on myeloid cells in the TME. CONCLUSIONS: Taken together, our data show that although treatment with cisplatin, topotecan and vaccination initially delays T cell expansion, this combinatorial therapy results eventually in a more robust T cell-mediated tumor eradication due to enhancement of costimulatory molecules in the TME.


Subject(s)
Cancer Vaccines , Uterine Cervical Neoplasms , Female , Humans , Animals , Mice , Cisplatin/pharmacology , Cisplatin/therapeutic use , CD8-Positive T-Lymphocytes , Cancer Vaccines/therapeutic use , Topotecan/pharmacology , Topotecan/therapeutic use , DNA Topoisomerases, Type I , Papillomavirus E7 Proteins , Vaccines, Subunit , Uterine Cervical Neoplasms/drug therapy , Cell Proliferation , Tumor Microenvironment , CD27 Ligand
2.
Front Immunol ; 14: 1163118, 2023.
Article in English | MEDLINE | ID: mdl-37781393

ABSTRACT

Introduction: Therapeutic vaccination based on synthetic long peptides (SLP®) containing both CD4+ and CD8+ T cell epitopes is a promising treatment strategy for chronic hepatitis B infection (cHBV). Methods: We designed SLPs for three HBV proteins, HBcAg and the non-secreted proteins polymerase and X, and investigated their ability to induce T cell responses ex vivo. A set of 17 SLPs was constructed based on viral protein conservation, functionality, predicted and validated binders for prevalent human leukocyte antigen (HLA) supertypes, validated HLA I epitopes, and chemical producibility. Results: All 17 SLPs were capable of inducing interferon gamma (IFNÉ£) production in samples from four or more donors that had resolved an HBV infection in the past (resolver). Further analysis of the best performing SLPs demonstrated activation of both CD8+ and CD4+ multi-functional T cells in one or more resolver and patient sample(s). When investigating which SLP could activate HBV-specific T cells, the responses could be traced back to different peptides for each patient or resolver. Discussion: This indicates that a large population of subjects with different HLA types can be covered by selecting a suitable mix of SLPs for therapeutic vaccine design. In conclusion, we designed a set of SLPs capable of inducing multifunctional CD8+ and CD4+ T cells ex vivo that create important components for a novel therapeutic vaccine to cure cHBV.


Subject(s)
CD4-Positive T-Lymphocytes , Hepatitis B virus , Humans , Interferon-gamma/metabolism , Histocompatibility Antigens Class I/metabolism , CD8-Positive T-Lymphocytes , Histocompatibility Antigens Class II/metabolism , Peptides , HLA Antigens/metabolism , Epitopes, T-Lymphocyte
3.
Clin Cancer Res ; 29(20): 4109-4117, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37540563

ABSTRACT

PURPOSE: Anal cancer is increasing in HIV+ men who have sex with men (MSM). Treatment options for its precursor, high-grade anal intraepithelial neoplasia (HGAIN), are suboptimal. In this phase I to II dose-finding study, we assessed the safety and efficacy of the human papillomavirus type 16 (HPV16) synthetic long peptide vaccine (SLP-HPV-01) in HIV+ MSM with HPV16-positive HGAIN. PATIENTS AND METHODS: Four dosage schedules (1-5-10; 5-10-20; 10-20-40; and 40-40-40-40 µg) of SLP-HPV-01 were administered intradermally with a 3-week interval in 10 patients per dose level (DL). In each dose group, 5 patients also received 1 µg/kg pegylated IFNα-2b subcutaneously. Primary endpoints were safety and regression of HGAIN at 3, 6, and 12 months. RESULTS: Eighty-one of 134 screened patients (60%) had HPV16-negative HGAIN lesions, leaving 53 eligible patients. Thirteen patients were excluded, leaving 40 men. The vaccine was well tolerated. One patient developed a generalized rash. The highest dosage level induced the strongest immune responses. There was no indication for stronger reactivity in the IFNα groups. Up to 18 months of follow-up, 8/38 intention-to-treat patients had a complete clinical and histologic response and one had a partial response (in total 9/38, 23.7%). At the highest dosage level, the clinical response was 4/10 (40%). Stronger immune responses were detected among clinical responders. CONCLUSIONS: The highest DL is safe, immunogenic, and associated with clinical responses to HPV16-induced lesions. However, as the majority of HGAIN is caused by the other HPV types, further studies should aim at pan-HPV vaccination to prevent or treat HGAIN.


Subject(s)
Anus Neoplasms , Cancer Vaccines , HIV Infections , Papillomavirus Infections , Sexual and Gender Minorities , Male , Humans , Homosexuality, Male , Human papillomavirus 16 , Human Papillomavirus Viruses , Papillomavirus Infections/complications , Papillomavirus Infections/prevention & control , Anus Neoplasms/pathology , Vaccination , Cancer Vaccines/adverse effects , HIV Infections/complications , HIV Infections/drug therapy
4.
Cancer Immunol Immunother ; 72(8): 2851-2864, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37222770

ABSTRACT

Therapeutic cancer vaccines trigger CD4 + and CD8 + T cell responses capable of established tumor eradication. Current platforms include DNA, mRNA and synthetic long peptide (SLP) vaccines, all aiming at robust T cell responses. SLPs linked to the Amplivant® adjuvant (Amplivant-SLP) have shown effective delivery to dendritic cells, resulting in improved immunogenicity in mice. We have now tested virosomes as a delivery vehicle for SLPs. Virosomes are nanoparticles made from influenza virus membranes and have been used as vaccines for a variety of antigens. Amplivant-SLP virosomes induced the expansion of more antigen-specific CD8 + T memory cells in ex vivo experiments with human PBMCs than Amplivant-SLP conjugates alone. The immune response could be further improved by including the adjuvants QS-21 and 3D-PHAD in the virosomal membrane. In these experiments, the SLPs were anchored in the membrane through the hydrophobic Amplivant adjuvant. In a therapeutic mouse model of HPV16 E6/E7+ cancer, mice were vaccinated with virosomes loaded with either Amplivant-conjugated SLPs or lipid-coupled SLPs. Vaccination with both types of virosomes significantly improved the control of tumor outgrowth, leading to elimination of the tumors in about half the animals for the best combinations of adjuvants and to their survival beyond 100 days.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Animals , Mice , Virosomes , Human papillomavirus 16 , Papillomavirus E7 Proteins , Neoplasms/drug therapy , Vaccination , Adjuvants, Immunologic , CD8-Positive T-Lymphocytes , Peptides , Vaccines, Synthetic , Mice, Inbred C57BL
5.
Semin Immunopathol ; 45(2): 273-277, 2023 03.
Article in English | MEDLINE | ID: mdl-36780000

ABSTRACT

Therapeutic vaccination of premalignant conditions and of different stages of cancer can be accomplished with several platforms including DNA vaccines, RNA vaccines, synthetic long peptides (SLP), and recombinant viruses. We successfully used a therapeutic vaccine composed of SLP covering the complete sequence of the two oncogenic proteins E6 and E7 of human papillomavirus type 16 (HPV16) as monotherapy in patients with premalignant disease. However, combination treatment might be required in patients with (advanced) cancer because of the hostile cancer microenvironment for T cells in established HPV16+ cancer, often associated with systemic immunosuppression. In patients with late-stage recurrent or metastatic HPV16+ cancers, we have therefore combined treatment with the SLP vaccine, called ISA101b, with either standard-of-care chemotherapy or with immune checkpoint inhibition with anti-PD-1 monoclonal antibody. A strong vaccine-induced interferon gamma-producing T cell response to HPV16 E6/E7 was associated with significantly better survival. In a second phase 1/2 study, patients with recurrent or metastatic HPV16+ oropharyngeal cancer were treated with the combination of ISA101b and anti-PD-1 (nivolumab). In this trial, the clinical overall response rate (ORR) in 22 patients was 36%, twice the ORR in the nivolumab registration trial for this category of patients, and 2/22 patients had a complete clinical response that is ongoing after 4 1/2 years. Other promising strategies for late-stage cancer recipients are the infusion of expanded tumor-infiltrating lymphocytes or the infusion of T cell receptor transduced T cells, both directed against HPV16.


Subject(s)
Cancer Vaccines , Neoplasms , Papillomavirus Infections , Humans , Cancer Vaccines/therapeutic use , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Human papillomavirus 16 , Human Papillomavirus Viruses , Immunotherapy , Neoplasms/drug therapy , Neoplasms/etiology , Nivolumab/pharmacology , Nivolumab/therapeutic use , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/drug therapy , Peptides , Tumor Microenvironment , Programmed Cell Death 1 Receptor/antagonists & inhibitors
6.
Cell Rep Med ; 3(10): 100788, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36260984

ABSTRACT

Padron et al.1 studied the combination of chemotherapy (gemcitabine and nab-paclitaxel) with either an anti-PD1 (nivolumab) or an anti-CD40 (sotigalimab) antibody in metastatic pancreatic cancer. They showed clinical benefit in individuals with unique biomarkers for each treatment combination.


Subject(s)
Nivolumab , Pancreatic Neoplasms , Humans , Nivolumab/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Treatment Outcome , Pancreatic Neoplasms/drug therapy , Biomarkers , Immunotherapy , Pancreatic Neoplasms
7.
J Immunother Cancer ; 10(10)2022 10.
Article in English | MEDLINE | ID: mdl-36261215

ABSTRACT

BACKGROUND: Amplivant is a molecularly optimized Toll-like receptor 2 ligand that can be covalently conjugated to tumor peptide antigens. In preclinical models, amplivant-adjuvanted synthetic long peptides (SLPs) strongly enhanced antigen presentation by dendritic cells, T cell priming and induction of effective antitumor responses. The current study is a first-in-human trial to investigate safety and immunogenicity of amplivant conjugated to human papillomavirus (HPV) 16-SLP. METHODS: A dose escalation phase I vaccination trial was performed in 25 patients treated for HPV16 positive (pre-)malignant lesions. Amplivant was conjugated to two SLPs derived from the two most immunodominant regions of the HPV16 E6 oncoprotein. The vaccine, containing a mix of these two conjugates in watery solution without any other formulation, was injected intradermally three times with a 3-week interval in four dose groups (1, 5, 20 or 50 µg per conjugated peptide). Safety data were collected during the study. Peptide-specific T cell immune responses were determined in blood samples taken before, during and after vaccination using complementary immunological assays. RESULTS: Toxicity after three amplivant-conjugated HPV16-SLP vaccinations was limited to grade 1 or 2, observed as predominantly mild skin inflammation at the vaccination site and sometimes mild flu-like symptoms. Adverse events varied from none in the lowest dose group to mild/moderate vaccine-related inflammation in all patients and flu-like symptoms in three out of seven patients in the highest dose group, after at least one injection. In the lowest dose group, vaccine-induced T cell responses were observed in the blood of three out of six vaccinated persons. In the highest dose group, all patients displayed a strong HPV16-specific T cell response after vaccination. These HPV16-specific T cell responses lasted until the end of the trial. CONCLUSIONS: Amplivant-conjugated SLPs can safely be used as an intradermal therapeutic vaccine to induce robust HPV16-specific T cell immunity in patients previously treated for HPV16 positive (pre-) malignancies. Increased vaccine dose was associated with a higher number of mild adverse events and with stronger systemic T cell immunity. TRIAL REGISTRATION NUMBERS: NCT02821494 and 2014-000658-12.


Subject(s)
Human papillomavirus 16 , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Female , Humans , Immunodominant Epitopes , Inflammation/etiology , Ligands , Peptides , T-Lymphocytes , Toll-Like Receptor 2 , Uterine Cervical Neoplasms/virology , Papillomavirus Vaccines/adverse effects , Papillomavirus Infections/complications
8.
Nat Commun ; 13(1): 3966, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35803932

ABSTRACT

Understanding the mechanisms and impact of booster vaccinations are essential in the design and delivery of vaccination programs. Here we show that a three dose regimen of a synthetic peptide vaccine elicits an accruing CD8+ T cell response against one SARS-CoV-2 Spike epitope. We see protection against lethal SARS-CoV-2 infection in the K18-hACE2 transgenic mouse model in the absence of neutralizing antibodies, but two dose approaches are insufficient to confer protection. The third vaccine dose of the single T cell epitope peptide results in superior generation of effector-memory T cells and tissue-resident memory T cells, and these tertiary vaccine-specific CD8+ T cells are characterized by enhanced polyfunctional cytokine production. Moreover, fate mapping shows that a substantial fraction of the tertiary CD8+ effector-memory T cells develop from re-migrated tissue-resident memory T cells. Thus, repeated booster vaccinations quantitatively and qualitatively improve the CD8+ T cell response leading to protection against otherwise lethal SARS-CoV-2 infection.


Subject(s)
COVID-19 , Epitopes, T-Lymphocyte , Animals , Antibodies, Neutralizing , Antibodies, Viral , CD8-Positive T-Lymphocytes , COVID-19/prevention & control , Disease Models, Animal , Immunologic Memory , Mice , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccination , Vaccines, Synthetic
10.
J Immunother Cancer ; 10(2)2022 02.
Article in English | MEDLINE | ID: mdl-35193933

ABSTRACT

BACKGROUND: The combination of ISA101, a human papilloma virus (HPV) 16 peptide vaccine, and nivolumab showed a promising response rate of 33% in patients with incurable HPV-16+ cancer. Here we report long-term clinical outcomes and immune correlates of response. METHODS: Patients with advanced HPV-16+ cancer and less than two prior regimens for recurrence were enrolled to receive ISA101 (100 µg/peptide) on days 1, 22, and 50 and nivolumab 3 mg/kg every 2 weeks beginning day 8 for up to 1 year. Baseline tumor samples were stained with multiplex immunofluorescence for programmed death-ligand 1 (PD-L1), programmed cell death protein-1 (PD-1), CD3, CD8, CD68, and pan-cytokeratin in a single panel and scanned with the Vectra 3.0 multispectral microscope. Whole transcriptome analysis of baseline tumors was performed with Affymetrix Clariom D arrays. Differential gene expression analysis was performed on responders versus non-responders. RESULTS: Twenty-four patients were followed for a median of 46.5 months (95% CI, 46.0 months to not reached (NR)). The median duration of response was 11.2 months (95% CI, 8.51 months to NR); three out of eight (38%) patients with objective response were without progression at 3 years. The median and 3-year overall survival were 15.3 months (95% CI, 10.6 months to 27.2 months) and 12.5% (95% CI, 4.3% to 36%), respectively. The scores for activated T cells ((CD3+PD-1+)+(CD3+CD8+PD-1+)), activated cytotoxic T cells (CD3+CD8+PD-1+), and total macrophage ((CD68+PD-L1-)+(CD68+PD-L1+)) in tumor were directly correlated with clinical response (p<0.05) and depth of response with the two complete response patients having the highest degree of CD8+ T cells. Gene expression analysis revealed differential regulation of 357 genes (≥1.25 fold) in non-responders versus responders (p<0.05). Higher expression of immune response, inflammatory response and interferon-signaling pathway genes were correlated with clinical response (p<0.05). CONCLUSIONS: Efficacy of ISA101 and nivolumab remains promising in long-term follow-up. Increased infiltration by PD-1+ T cells and macrophages was predictive of response. Enrichment in gene sets associated with interferon-γ response and immune infiltration strongly predicted response to therapy. A randomized trial is ongoing to test this strategy and to further explore correlates of immune response with combined nivolumab and ISA101, versus nivolumab alone. TRIAL REGISTRATION NUMBER: NCT02426892.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Human papillomavirus 16/drug effects , Human papillomavirus 16/immunology , Immunity/immunology , Nivolumab/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Female , Humans , Male , Nivolumab/pharmacology
11.
J Immunother Cancer ; 10(9)2022 09.
Article in English | MEDLINE | ID: mdl-36939214

ABSTRACT

Water-in-oil emulsion incomplete Freund's adjuvant (IFA) has been used as an adjuvant in preventive and therapeutic vaccines since its development. New generation, highly purified modulations of the adjuvant, Montanide incomplete seppic adjuvant (ISA)-51 and Montanide ISA-720, were developed to reduce toxicity. Montanide adjuvants are generally considered to be safe, with adverse events largely consisting of antigen and adjuvant dose-dependent injection site reactions (ISRs). Peptide vaccines in Montanide ISA-51 or ISA-720 are capable of inducing both high antibody titers and durable effector T cell responses. However, an efficient T cell response depends on the affinity of the peptide to the presenting major histocompatibility complex class I molecule, CD4+ T cell help and/or the level of co-stimulation. In fact, in the therapeutic cancer vaccine setting, presence of a CD4+ T cell epitope seems crucial to elicit a robust and durable systemic T cell response. Additional inclusion of a Toll-like receptor ligand can further increase the magnitude and durability of the response. Use of extended peptides that need a processing step only accomplished effectively by dendritic cells (DCs) can help to avoid antigen presentation by nucleated cells other than DC. Based on recent clinical trial results, therapeutic peptide-based cancer vaccines using emulsions in adjuvant Montanide ISA-51 can elicit robust antitumor immune responses, provided that sufficient tumor-specific CD4+ T cell help is given in addition to CD8+ T cell epitopes. Co-treatment with PD-1 T cell checkpoint inhibitor, chemotherapy or other immunomodulatory drugs may address local and systemic immunosuppressive mechanisms, and further enhance efficacy of therapeutic cancer peptide vaccines in IFA and its modern variants. Blinded randomized placebo-controlled trials are critical to definitively prove clinical efficacy. Mineral oil-based adjuvants for preventive vaccines, to tackle spread and severity of infectious disease, induce immune responses, but require more studies to reduce toxicity.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Adjuvants, Immunologic , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Emulsions , Immunotherapy , Neoplasms/drug therapy , Peptides , Vaccines, Subunit
12.
Exp Dermatol ; 31(4): 556-566, 2022 04.
Article in English | MEDLINE | ID: mdl-34758170

ABSTRACT

Mounting evidence shows that the PD-1/PD-L1 axis is involved in tumor immune evasion. This is demonstrated by anti-PD-1 antibodies that can reverse tumor-associated PD-L1 to functionally suppress anti-tumor T-cell responses. Since type I and II interferons are key regulators of PD-L1 expression in melanoma cells and IFN-γ-producing CD8+ T cells and IFN-α-producing dendritic cells are abundant in vitiligo skin, we aimed to study the role of PD-1/PD-L1 signalling in melanocyte destruction in vitiligo. Moreover, impaired PD-1/PD-L1 function is observed in a variety of autoimmune diseases. It is, therefore, hypothesized that manipulating PD-1/PD-L1 signalling might have therapeutic potential in vitiligo. The PD-1+ T cells were abundantly present in situ in perilesional vitiligo skin, but expression of PD-L1 was limited and confined exclusively to dermal T cells. More specifically, neither melanocytes nor other epidermal skin cells expressed PD-L1. Exposure to IFN-γ, but also type I interferons, increased PD-L1 expression in primary melanocytes and fibroblasts, derived from healthy donors. Primary human keratinocytes only showed increased PD-L1 expression upon stimulation with IFN-γ. More interestingly, melanocytes derived from non-lesional vitiligo skin showed no PD-L1 upregulation upon IFN-γ exposure, while other skin cells displayed significant PD-L1 expression after exposure. In a vitiligo skin explant model, incubation of non-lesional vitiligo skin with activated (IFN-γ-producing) T cells from vitiligo lesions was previously described to induce melanocyte apoptosis. Although PD-L1 expression was induced in epidermal cells in these explants, this induction was completely absent in melanocytes. The lack of PD-L1 upregulation by melanocytes in the presence of IFN-γ-producing T cells shows that melanocytes lack protection against T-cell attack during vitiligo pathogenesis. Manipulating PD-1/PD-L1 signalling may, therefore, be a therapeutic option for vitiligo patients.


Subject(s)
Vitiligo , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes , Humans , Interferon-gamma/metabolism , Melanocytes/metabolism , Programmed Cell Death 1 Receptor/metabolism
13.
Nat Rev Cancer ; 21(6): 360-378, 2021 06.
Article in English | MEDLINE | ID: mdl-33907315

ABSTRACT

Therapeutic cancer vaccines have undergone a resurgence in the past decade. A better understanding of the breadth of tumour-associated antigens, the native immune response and development of novel technologies for antigen delivery has facilitated improved vaccine design. The goal of therapeutic cancer vaccines is to induce tumour regression, eradicate minimal residual disease, establish lasting antitumour memory and avoid non-specific or adverse reactions. However, tumour-induced immunosuppression and immunoresistance pose significant challenges to achieving this goal. In this Review, we deliberate on how to improve and expand the antigen repertoire for vaccines, consider developments in vaccine platforms and explore antigen-agnostic in situ vaccines. Furthermore, we summarize the reasons for failure of cancer vaccines in the past and provide an overview of various mechanisms of resistance posed by the tumour. Finally, we propose strategies for combining suitable vaccine platforms with novel immunomodulatory approaches and standard-of-care treatments for overcoming tumour resistance and enhancing clinical efficacy.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Immune Tolerance , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/prevention & control , Tumor Microenvironment/immunology , Animals , Cancer Vaccines/immunology , Humans
14.
Cancer Immunol Res ; 9(5): 583-597, 2021 05.
Article in English | MEDLINE | ID: mdl-33619119

ABSTRACT

CD8+ T-cell infiltration and effector activity in tumors are correlated with better overall survival of patients, suggesting that the ability of T cells to enter and remain in contact with tumor cells supports tumor control. CD8+ T cells express the collagen-binding integrins CD49a and CD49b, but little is known about their function or how their expression is regulated in the tumor microenvironment (TME). Here, we found that tumor-infiltrating CD8+ T cells initially expressed CD49b, gained CD49a, and then lost CD49b over the course of tumor outgrowth. This differentiation sequence was driven by antigen-independent elements in the TME, although T-cell receptor (TCR) stimulation further increased CD49a expression. Expression of exhaustion markers and CD49a associated temporally but not mechanistically. Intratumoral CD49a-expressing CD8+ T cells failed to upregulate TCR-dependent Nur77 expression, whereas CD69 was constitutively expressed, consistent with both a lack of productive antigen engagement and a tissue-resident memory-like phenotype. Imaging T cells in live tumor slices revealed that CD49a increased their motility, especially of those in close proximity to tumor cells, suggesting that it may interfere with T-cell recognition of tumor cells by distracting them from productive engagement, although we were not able to augment productive engagement by short-term CD49a blockade. CD49b also promoted relocalization of T cells at a greater distance from tumor cells. Thus, our results demonstrate that expression of these integrins affects T-cell trafficking and localization in tumors via distinct mechanisms, and suggests a new way in which the TME, and likely collagen, could promote tumor-infiltrating CD8+ T-cell dysfunction.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/genetics , Integrin alpha1/metabolism , Integrin alpha2/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Humans , Male , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Tumor Microenvironment
15.
Immunother Adv ; 1(1): ltaa005, 2021 Jan.
Article in English | MEDLINE | ID: mdl-36756002

ABSTRACT

During the last two decades, two main schools of modern immunotherapy have come to the forefront. The chimeric anti-CD20 antibody rituximab that was introduced for the treatment of refractory follicular lymphoma in 1998 was one of the first examples of the school of passive immunotherapy. Subsequently major and ever more costly efforts were spent on the development of blockbuster monotherapies including other monoclonal but also bispecific antibodies of highly defined specificity and subclass, antibody-drug conjugates (ADCs), as well as ex vivo expanded tumor-infiltrating lymphocytes, chimeric antigen receptor (CAR)-transduced T cells, and TCR-transduced T cells. On the other hand, there is the school that works toward active induction of patient B- or T-cell immunity against antigens of choice, or active tolerance against pathogenic allergens, auto-antigens or allo-antigens. Stradled in between these two approaches is treatment with blockers of T cell checkpoint control, which releases the brakes of T cells that have already responded to antigen. Extensive and detailed insight into the cellular and molecular interactions that regulate specific immune responses is indispensable in order to be able to optimize efficacy and rule out treatment related toxicity. This applies to all types of immunotherapy. Our knowledge of the checks and balances in the immune system is still increasing at an unprecedented pace, fostering ever more effective and specific (combination) immunotherapies and offering a rich harvest of innovative immunotherapies in the years ahead.

16.
Front Immunol ; 11: 579022, 2020.
Article in English | MEDLINE | ID: mdl-33240267

ABSTRACT

Autoreactive CD8+ T cells play a pivotal role in melanocyte destruction in autoimmune vitiligo. Immunotherapy for melanoma often leads to autoimmune side-effects, among which vitiligo-like depigmentation, indicating that targeting immune checkpoints can break peripheral tolerance against self-antigens in the skin. Therapeutically enhancing immune checkpoint signaling by immune cells or skin cells, making self-reactive T cells anergic, seems a promising therapeutic option for vitiligo. Here, we review the current knowledge on the PD-1/PD-L1 pathway in vitiligo as new therapeutic target for vitiligo therapy.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Melanocytes/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Pigmentation/drug effects , Skin/drug effects , Vitiligo/drug therapy , Animals , Autoimmunity/drug effects , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Melanocytes/immunology , Melanocytes/metabolism , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Skin/immunology , Skin/metabolism , Skin/physiopathology , Vitiligo/immunology , Vitiligo/metabolism , Vitiligo/physiopathology
17.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32561639

ABSTRACT

BACKGROUND: Stress-induced post-translational modifications occur during autophagy and can result in generation of new epitopes and immune recognition. One such modification is the conversion of arginine to citrulline by peptidylarginine deiminase enzymes. METHODS: We used Human leukocyte antigen (HLA) transgenic mouse models to assess the immunogenicity of citrullinated peptide vaccine by cytokine Enzyme linked immunosorbant spot (ELISpot) assay. Vaccine efficacy was assessed in tumor therapy studies using HLA-matched B16 melanoma and ID8 ovarian models expressing either constitutive or interferon-gamma (IFNγ) inducible Major Histocompatibility Complex (MHC) class II (MHC-II) as represented by most human tumors. To determine the importance of CD4 T cells in tumor therapy, we analyzed the immune cell infiltrate into murine tumors using flow cytometry and performed therapy studies in the presence of CD4 and CD8 T cell depletion. We assessed the T cell repertoire to citrullinated peptides in ovarian cancer patients and healthy donors using flow cytometry. RESULTS: The combination of citrullinated vimentin and enolase peptides (Modi-1) stimulated strong CD4 T cell responses in mice. Responses resulted in a potent anti-tumor therapy against established tumors and generated immunological memory which protected against tumor rechallenge. Depletion of CD4, but not CD8 T cells, abrogated the primary anti-tumor response as well as the memory response to tumor rechallenge. This was further reinforced by successful tumor regression being associated with an increase in tumor-infiltrating CD4 T cells and a reduction in tumor-associated myeloid suppressor cells. The anti-tumor response also relied on direct CD4 T cell recognition as only tumors expressing MHC-II were rejected. A comparison of different Toll-like receptor (TLR)-stimulating adjuvants showed that Modi-1 induced strong Th1 responses when combined with granulocyte-macrophage colony-stimulating factor (GMCSF), TLR9/TLR4, TLR9, TLR3, TLR1/2 and TLR7 agonists. Direct linkage of the TLR1/2 agonist to the peptides allowed the vaccine dose to be reduced by 10-fold to 100-fold without loss of anti-tumor activity. Furthermore, a CD4 Th1 response to the citrullinated peptides was seen in ovarian cancer patients. CONCLUSIONS: Modi-1 citrullinated peptide vaccine induces potent CD4-mediated anti-tumor responses in mouse models and a CD4 T cell repertoire is present in ovarian cancer patients to the citrullinated peptides suggesting that Modi-1 could be an effective vaccine for ovarian cancer patients.


Subject(s)
Cancer Vaccines/immunology , Melanoma, Experimental/therapy , Phosphopyruvate Hydratase/immunology , Vimentin/immunology , Adjuvants, Immunologic/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Citrullination/immunology , Female , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Immunogenicity, Vaccine , Interferon-gamma/immunology , Lymphocyte Depletion , Male , Melanoma, Experimental/immunology , Mice , Mice, Transgenic , Phosphopyruvate Hydratase/genetics , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vimentin/genetics
18.
Sci Transl Med ; 12(535)2020 03 18.
Article in English | MEDLINE | ID: mdl-32188726

ABSTRACT

Therapeutic cancer vaccines have effectively induced durable regressions of premalignant oncogenic human papilloma virus type 16 (HPV16)-induced anogenital lesions. However, the treatment of HPV16-induced cancers requires appropriate countermeasures to overcome cancer-induced immune suppression. We previously showed that standard-of-care carboplatin/paclitaxel chemotherapy can reduce abnormally high numbers of immunosuppressive myeloid cells in patients, allowing the development of much stronger therapeutic HPV16 vaccine (ISA101)-induced tumor immunity. We now show the clinical effects of ISA101 vaccination during chemotherapy in 77 patients with advanced, recurrent, or metastatic cervical cancer in a dose assessment study of ISA101. Tumor regressions were observed in 43% of 72 evaluable patients. The depletion of myeloid suppressive cells by carboplatin/paclitaxel was associated with detection of low frequency of spontaneous HPV16-specific immunity in 21 of 62 tested patients. Patients mounted type 1 T cell responses to the vaccine across all doses. The group of patients with higher than median vaccine-induced immune responses lived longer, with a flat tail on the survival curve. This demonstrates that chemoimmunotherapy can be exploited to the benefit of patients with advanced cancer based on a defined mode of action.


Subject(s)
Cancer Vaccines , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Female , Human papillomavirus 16 , Humans , Papillomavirus E7 Proteins
19.
Antiviral Res ; 178: 104746, 2020 06.
Article in English | MEDLINE | ID: mdl-32081741

ABSTRACT

Synthetic long peptide (SLP) vaccination is a promising new treatment strategy for patients with a chronic hepatitis B virus (HBV) infection. We have previously shown that a prototype HBV-core protein derived SLP was capable of boosting CD4+ and CD8+ T cell responses in the presence of a TLR2-ligand in chronic HBV patients ex vivo. For optimal efficacy of a therapeutic vaccine in vivo, adjuvants can be conjugated to the SLP to ensure delivery of both the antigen and the co-stimulatory signal to the same antigen-presenting cell (APC). Dendritic cells (DCs) express the receptor for the adjuvant and are optimally equipped to efficiently process and present the SLP-contained epitopes to T cells. Here, we investigated TLR2-ligand conjugation of the prototype HBV-core SLP. Results indicated that TLR2-ligand conjugation reduced cross-presentation efficiency of the SLP-contained epitope by both monocyte-derived and naturally occurring DC subsets. Importantly, cross-presentation was improved after optimization of the conjugate by either shortening the SLP or by placing a valine-citrulline linker between the TLR2-ligand and the long SLP, to facilitate endosomal dissociation of SLP and TLR2-ligand after uptake. HBV-core SLP conjugates also triggered functional patient T cell responses ex vivo. These results provide an import step forward in the design of a therapeutic SLP-based vaccine to cure chronic HBV.


Subject(s)
Hepatitis B Vaccines/therapeutic use , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/therapy , T-Lymphocytes/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Adjuvants, Immunologic , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Dendritic Cells/immunology , Epitopes, T-Lymphocyte , Hepatitis B Core Antigens/immunology , Hepatitis B Core Antigens/metabolism , Hepatitis B Vaccines/immunology , Humans , Ligands , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
20.
J Immunother Cancer ; 7(1): 10, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30646957

ABSTRACT

BACKGROUND: Chemoradiotherapy (CRT) remains one of the most common cancer treatment modalities, and recent data suggest that CRT is maximally effective when there is generation of an anti-tumoral immune response. However, CRT has also been shown to promote immunosuppressive mechanisms which must be blocked or reversed to maximize its immune stimulating effects. METHODS: Therefore, using a preclinical model of human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC), we developed a clinically relevant therapy combining CRT and two existing immunomodulatory drugs: cyclophosphamide (CTX) and the small molecule inducible nitric oxide synthase (iNOS) inhibitor L-n6-(1-iminoethyl)-lysine (L-NIL). In this model, we treated the syngeneic HPV-HNSCC mEER tumor-bearing mice with fractionated (10 fractions of 3 Gy) tumor-directed radiation and weekly cisplatin administration. We compared the immune responses induced by CRT and those induced by combinatory treatment (CRT + CTX/L-NIL) with flow cytometry, quantitative multiplex immunofluorescence and by profiling immune-related gene expression changes. RESULTS: We show that combination treatment favorably remodels the tumor myeloid immune microenvironment including an increase in anti-tumor immune cell types (inflammatory monocytes and M1-like macrophages) and a decrease in immunosuppressive granulocytic myeloid-derived suppressor cells (MDSCs). Intratumoral T cell infiltration and tumor antigen specificity of T cells were also improved, including a 31.8-fold increase in the CD8+ T cell/ regulatory T cell ratio and a significant increase in tumor antigen-specific CD8+ T cells compared to CRT alone. CTX/LNIL immunomodulation was also shown to significantly improve CRT efficacy, leading to rejection of 21% established tumors in a CD8-dependent manner. CONCLUSIONS: Overall, these data show that modulation of the tumor immune microenvironment with CTX/L-NIL enhances susceptibility of treatment-refractory tumors to CRT. The combination of tumor immune microenvironment modulation with CRT constitutes a translationally relevant approach to enhance CRT efficacy through enhanced immune activation.


Subject(s)
Antineoplastic Agents/therapeutic use , Chemoradiotherapy , Cyclophosphamide/therapeutic use , Head and Neck Neoplasms/therapy , Immunomodulation , Lysine/analogs & derivatives , Neoplasms/therapy , Squamous Cell Carcinoma of Head and Neck/therapy , Tumor Microenvironment/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Head and Neck Neoplasms/immunology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lysine/therapeutic use , Male , Mice, Inbred C57BL , Papillomavirus Infections/complications , Squamous Cell Carcinoma of Head and Neck/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...