Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Mol Med ; 28(1): 37, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35317721

ABSTRACT

BACKGROUND: Chronic liver injury induces pathological repair, resulting in fibrosis, during which hepatic stellate cells (HSCs) are activated and transform into myofibroblasts. CD248 is mainly expressed on myofibroblasts and was considered as a promising target to treat fibrosis. The primary aim of this study was to generate a CD248 specific antibody-drug conjugate (ADC) and evaluate its therapeutic efficacy for liver fibrosis and its safety in vivo. METHODS: CD248 expression was examined in patients with liver cirrhosis and in mice with CCl4-induced liver fibrosis. The ADC IgG78-DM1, which targets CD248, was prepared and its bioactivity on activated primary HSCs was studied. The anti-fibrotic effects of IgG78-DM1 on liver fibrosis were evaluated in CCl4-induced mice. The reproductive safety and biosafety of IgG78-DM1 were also evaluated in vivo. RESULTS: CD248 expression was upregulated in patients with liver cirrhosis and in CCl4-induced mice, and was mainly expressed on alpha smooth muscle actin (α-SMA)+ myofibroblasts. IgG78-DM1 was successfully generated, which could effectively bind with and kill CD248+ activated HSCs in vitro and inhibit liver fibrosis in vivo. In addition, IgG78-DM1 was demonstrated to have qualified biosafety and reproductive safety in vivo. CONCLUSIONS: Our study demonstrated that CD248 could be an ideal target for myofibroblasts in liver fibrosis, and CD248-targeting IgG78-DM1 had excellent anti-fibrotic effects in mice with liver fibrosis. Our study provided a novel strategy to treat liver fibrosis and expanded the application of ADCs beyond tumors.


Subject(s)
Immunoconjugates , Myofibroblasts , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/metabolism , Fibrosis , Hepatic Stellate Cells/metabolism , Humans , Immunoconjugates/adverse effects , Immunoconjugates/metabolism , Liver/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Mice , Myofibroblasts/metabolism
2.
J Immunother Cancer ; 9(7)2021 07.
Article in English | MEDLINE | ID: mdl-34233963

ABSTRACT

With durable cancer responses, genetically modified cell therapies are being implemented in various cancers. However, these immune effector cell therapies can cause toxicities, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Pseudogout arthritis is an inflammatory arthritis induced by deposition of calcium pyrophosphate dihydrate crystals. Here, we report a case of pseudogout arthritis in a patient treated with MAGE-A4 directed T cell receptor T cells, for fallopian tube cancer. The patient developed CRS and ICANS 7 days after infusion of the T cells. Concurrently, the patient newly developed sudden onset of left knee arthritis. Synovial fluid analyses revealed the presence of calcium pyrophosphate dihydrate crystal. Notably, the pseudogout arthritis was resolved with tocilizumab, which was administered for the treatment of CRS and ICANS. Immunoprofiling of the synovial fluid showed that the proportion of inflammatory interleukin 17 (IL-17)-producing CD4+ T (Th17) cells and amount of IL-6 were notably increased, suggesting a potential role of Th17 cells in pseudogout arthritis after T-cell therapy. To the best of our knowledge, this is the first reported case of pseudogout arthritis after cell therapy. Clinicians, especially hematologists, oncologists and rheumatologists, should be aware that pseudogout arthritis can be associated with CRS/ICANS.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Neoplasm/adverse effects , Chondrocalcinosis/etiology , Neoplasm Proteins/adverse effects , Receptors, Antigen, T-Cell/therapeutic use , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Chondrocalcinosis/physiopathology , Female , Humans
3.
Front Immunol ; 12: 623639, 2021.
Article in English | MEDLINE | ID: mdl-33692796

ABSTRACT

Although melanoma remains the deadliest skin cancer, the current treatment has not resulted in the desired outcomes. Unlike chemotherapy, immunotherapy has provided more tolerable approaches and revolutionized cancer therapy. Although dendritic cell-based vaccines have minor side effects, the undesirable response rates of traditional approaches have posed questions about their clinical translation. The immunosuppressive tumor microenvironment can be the underlying reason for their low response rates. Immune checkpoints and indoleamine 2,3-dioxygenase have been implicated in the induction of immunosuppressive tumor microenvironment. Growing evidence indicates that the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase/Protein kinase B (PKB) (PI3K/AKT) pathways, as the main oncogenic pathways of melanoma, can upregulate the tumoral immune checkpoints, like programmed death-ligand 1. This study briefly represents the main oncogenic pathways of melanoma and highlights the cross-talk between these oncogenic pathways with indoleamine 2,3-dioxygenase, tumoral immune checkpoints, and myeloid-derived suppressor cells. Moreover, this study sheds light on a novel tumor antigen on melanoma, which has substantial roles in tumoral immune checkpoints expression, indoleamine 2,3-dioxygenase secretion, and stimulating the oncogenic pathways. Finally, this review collects the lessons from the previous unsuccessful trials and integrates their lessons with new approaches in RNA-modified dendritic cell vaccines. Unlike traditional approaches, the advances in single-cell RNA-sequencing techniques and RNA-modified dendritic cell vaccines along with combined therapy of the immune checkpoint inhibitors, indoleamine 2,3-dioxygenase inhibitor, and RNA-modified dendritic cell-based vaccine can overcome these auto-inductive loops and pave the way for developing robust dendritic cell-based vaccines with the most favorable response rate and the least side effects.


Subject(s)
Antigens, Neoplasm/therapeutic use , Cancer Vaccines/therapeutic use , Dendritic Cells/transplantation , Melanoma/therapy , RNA, Small Interfering/therapeutic use , Skin Neoplasms/therapy , Vaccines, Synthetic/therapeutic use , Animals , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immune Checkpoint Proteins/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , RNA, Small Interfering/adverse effects , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Tumor Escape , Tumor Microenvironment , Vaccines, Synthetic/adverse effects , mRNA Vaccines
4.
Clin Cancer Res ; 26(17): 4503-4510, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32540851

ABSTRACT

PURPOSE: DNA mismatch repair (MMR) deficiency is a hallmark of Lynch syndrome, the most common inherited cancer syndrome. MMR-deficient cancer cells accumulate numerous insertion/deletion mutations at microsatellites. Mutations of coding microsatellites (cMS) lead to the generation of immunogenic frameshift peptide (FSP) neoantigens. As the evolution of MMR-deficient cancers is triggered by mutations inactivating defined cMS-containing tumor suppressor genes, distinct FSP neoantigens are shared by most MMR-deficient cancers. To evaluate safety and immunogenicity of an FSP-based vaccine, we performed a clinical phase I/IIa trial (Micoryx). PATIENTS AND METHODS: The trial comprised three cycles of four subcutaneous vaccinations (FSP neoantigens derived from mutant AIM2, HT001, TAF1B genes) mixed with Montanide ISA-51 VG over 6 months. Inclusion criteria were history of MMR-deficient colorectal cancer (UICC stage III or IV) and completion of chemotherapy. Phase I evaluated safety and toxicity as primary endpoint (six patients), phase IIa addressed cellular and humoral immune responses (16 patients). RESULTS: Vaccine-induced humoral and cellular immune responses were observed in all patients vaccinated per protocol. Three patients developed grade 2 local injection site reactions. No vaccination-induced severe adverse events occurred. One heavily pretreated patient with bulky metastases showed stable disease and stable CEA levels over 7 months. CONCLUSIONS: FSP neoantigen vaccination is systemically well tolerated and consistently induces humoral and cellular immune responses, thus representing a promising novel approach for treatment and even prevention of MMR-deficient cancer.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Colorectal Neoplasms, Hereditary Nonpolyposis/therapy , Colorectal Neoplasms/prevention & control , DNA Mismatch Repair , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/genetics , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms, Hereditary Nonpolyposis/complications , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/immunology , DNA-Binding Proteins/genetics , Female , Frameshift Mutation , Humans , Injections, Subcutaneous , Male , Microsatellite Instability , Middle Aged , Pol1 Transcription Initiation Complex Proteins/genetics , Proteins/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , Vaccines, Subunit/genetics
5.
Int J Biol Markers ; 34(3): 251-261, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31436122

ABSTRACT

OBJECTIVE: The aim of the study was to evaluate the diagnostic value of soluble fragment of cytokeratin 19 (CYFRA21-1) tests in detecting non-small cell lung cancer (NSCLC), including squamous cell carcinoma, lung adenocarcinoma, and large cell carcinoma. METHODS: The relevant studies were identified from PubMed, Embase and the Cochrane Library before November 2018. Summary estimates for sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio of CYFRA21-1 tests for the diagnosis of NSCLC were calculated using the random effects model. A summary receiver operating characteristic (SROC) curve was used to assess the overall effectiveness of the test. Meta-DiSc 1.4 and Stata11.0 were applied to the statistical analysis. Publication bias was detected using Egger's test. RESULTS: A total of 22 studies consisting of 7910 NSCLC patients (squamous cell carcinoma/lung adenocarcinoma/large cell carcinoma) and 2630 benign lesions patients that met the inclusion criteria were included. The meta-analysis showed that CYFRA21-1 tests had a relatively high accuracy for squamous cell carcinoma detection and a lower accuracy for lung adenocarcinoma detection. The overall sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio of CYFRA21-1 tests for squamous cell carcinoma detection were 0.72 (95% confidence interval (CI) 0.70, 0.74), 0.94 (95% CI 0.92, 0.95), 9.73 (95% CI 7.06, 13.40), 0.37 (95% CI 0.29, 0.47), and 27.30 (95% CI 17.68, 42.16), respectively. The area under the SROC curve was 0.9171 (Q* = 0.8500). No publication bias was tested in the squamous cell carcinoma (P = 0.567) and lung adenocarcinoma (P = 0.378) groups. CONCLUSIONS: CYFRA21-1 tests might be appropriate for detecting squamous cell carcinoma.


Subject(s)
Adenocarcinoma of Lung/diagnosis , Antigens, Neoplasm/adverse effects , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Squamous Cell/diagnosis , Keratin-19/adverse effects , Lung Neoplasms/diagnosis , Adenocarcinoma of Lung/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Humans , Lung Neoplasms/pathology
6.
Cancer Immunol Immunother ; 68(7): 1211-1222, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31069460

ABSTRACT

Human tumor cells express antigens that serve as targets for the host cellular immune system. This phase 1 dose-escalating study was conducted to assess safety and tolerability of G305, a recombinant NY-ESO-1 protein vaccine mixed with glucopyranosyl lipid A (GLA), a synthetic TLR4 agonist adjuvant, in a stable emulsion (SE). Twelve patients with solid tumors expressing NY-ESO-1 were treated using a 3 + 3 design. The NY-ESO-1 dose was fixed at 250 µg, while GLA-SE was increased from 2 to 10 µg. Safety, immunogenicity, and clinical responses were assessed prior to, during, and at the end of therapy. G305 was safe and immunogenic at all doses. All related AEs were Grade 1 or 2, with injection site soreness as the most commonly reported event (100%). Overall, 75% of patients developed antibody response to NY-ESO-1, including six patients with increased antibody titer ( ≥ 4-fold rise) and three patients with seroconversion from negative (titer < 100) to positive (titer ≥ 100). CD4 T-cell responses were observed in 44.4% of patients; 33.3% were new responses and 1 was boosted ( ≥ 2-fold rise). Following treatment, 8 of 12 patients had stable disease for 3 months or more; at the end of 1 year, three patients had stable disease and nine patients were alive. G305 is a potent immunotherapeutic agent that can stimulate NY-ESO-1-specific antibody and T-cell responses. The vaccine was safe at all doses of GLA-SE (2-10 µg) and showed potential clinical benefit in this population of patients.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Glucosides/administration & dosage , Lipid A/administration & dosage , Membrane Proteins/administration & dosage , Neoplasms/therapy , Adjuvants, Immunologic/adverse effects , Adult , Aged , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Female , Glucosides/adverse effects , Glucosides/immunology , Humans , Immunogenicity, Vaccine , Injections, Intramuscular , Lipid A/adverse effects , Lipid A/immunology , Male , Membrane Proteins/adverse effects , Membrane Proteins/immunology , Middle Aged , Neoplasms/immunology , Neoplasms/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Young Adult
7.
J Am Board Fam Med ; 31(4): 620-627, 2018.
Article in English | MEDLINE | ID: mdl-29986988

ABSTRACT

BACKGROUND: Immunotherapies in cancer treatment have a long history going all the way back to the very beginning of the field, and recent advances are extremely promising. These therapies are becoming a larger part in many patients' oncology treatment as the number of approaches, individual medicines, and indications increase. Furthermore, these novel therapies have different side effect profiles from those traditional chemotherapies which have, until recently, typified the oncologist's approach to treatment together with surgery and radiation. METHODS: An electronic literature search was conducted in May and June 2017 and March 2018 with the PubMed and Ebscohost databases. Articles were chosen for their relevance to the drugs in question, cancer physiology, or historic significance. CONCLUSIONS: Checkpoint inhibitors are becoming very common and possess autoimmune side effects such as pneumonitis, hypothyroidism, and colitis. These may present at any time the patient is on the medications but are more common several weeks to several months from beginning therapy. Chimeric antigen receptor T-cell therapies are powerful but have strong side effects such as cytokine release syndrome. Neoantigens are currently in the early stages of clinical trials and may become an exciting avenue for personalized cancer treatment but are not yet typical.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Costimulatory and Inhibitory T-Cell Receptors/antagonists & inhibitors , Immunotherapy, Adoptive/adverse effects , Neoplasms/therapy , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/adverse effects , Antineoplastic Agents, Immunological/administration & dosage , Clinical Trials as Topic , Colitis/epidemiology , Colitis/immunology , Costimulatory and Inhibitory T-Cell Receptors/immunology , Cytokines/immunology , Cytokines/metabolism , Humans , Hypothyroidism/epidemiology , Hypothyroidism/immunology , Immune System Diseases/epidemiology , Immune System Diseases/immunology , Neoplasms/immunology , Pneumonia/epidemiology , Pneumonia/immunology , Receptors, Chimeric Antigen/immunology , Treatment Outcome
8.
Vaccine ; 35(19): 2550-2557, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28385609

ABSTRACT

The lipid calcium phosphate nanoparticle is a versatile platform capable of encapsulating a wide range of phosphorylated molecules from single nucleotides to pDNA. The use of this platform has shown great success as an immunotherapeutic vaccine carrier, capable of delivering co-encapsulated phosphorylated adjuvants and peptides. Three potent vaccine formulations were investigated for anti-cancer efficacy. The phosphorylated adjuvants, CpG, 2'3'cGAMP, and 5'pppdsRNA were co-encapsulated with a model phosphorylated tumor specific peptide antigen (p-AH1-A5). The anti-cancer efficacy of these adjuvants was assessed using an orthotopic colorectal liver metastasis model based on highly aggressive and metastatic CT-26 FL3 cells implanted into the cecum wall. The results clearly indicate that the RIG-1 ligand, 5'pppdsRNA, co-encapsulated with the p-AH1-A5 peptide antigen greatly reduced the growth rate of the primary colon cancer as well as arrested the establishment of liver metastasis in comparison to the other adjuvant formulations and unvaccinated controls. Further evaluation of the immune cell populations within the primary tumor confirms the ability of the 5'pppdsRNA adjuvant to boost the adaptive CD8+ T-cell population, while not inciting increased populations of immune suppressive cell types such as T-regulatory cells or myeloid derived suppressor cells. Furthermore, to our knowledge this is the first study to investigate the anti-cancer efficacy of a specific RIG-1 receptor ligand, 5'pppdsRNA, alongside more established TLR 9 (CpG) and STING (2'3'cGAMP) adjuvants in a cancer vaccine. The 5'pppdsRNA vaccine formulation can be a potent immunotherapy, especially when combined with agents that remodel the immune suppressive microenvironment of the tumor.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Colorectal Neoplasms/complications , Colorectal Neoplasms/therapy , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Adaptive Immunity , Adjuvants, Immunologic/adverse effects , Animals , Antigens, Neoplasm/adverse effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/adverse effects , Colorectal Neoplasms/pathology , Disease Models, Animal , Female , Liver Neoplasms/pathology , Mice, Inbred BALB C , Neoplasm Metastasis/therapy , Peptides/administration & dosage , Peptides/adverse effects , Treatment Outcome
9.
Invest New Drugs ; 35(3): 315-323, 2017 06.
Article in English | MEDLINE | ID: mdl-28070718

ABSTRACT

Background The antibody-drug conjugate PF-06263507 targets the cell-surface, tumor-associated antigen 5T4 and consists of a humanized IgG1 conjugated to the microtubule-disrupting agent monomethylauristatin-F by a non-cleavable maleimidocaproyl linker. In this first-in-human, dose-finding trial (NCT01891669), we evaluated safety, pharmacokinetics, and preliminary antitumor activity of PF-06263507 in pretreated patients with advanced solid tumors, unselected for 5T4 expression. starting at 0.05 mg/kg, with 25, 56, and 95% dose increments, depending on observed dose-limiting toxicities (DLTs), applying a modified continual reassessment method. Results Twenty-six patients received PF-06263507 at 0.05 to 6.5 mg/kg. The first DLT, grade 3 photophobia, occurred at 4.34 mg/kg and two additional DLTs, grade 2 keratitis and grade 1 limbal stem cell deficiency (> 2-week dosing delay), at 6.5 mg/kg. The most common adverse events (AEs) were fatigue (38.5%), photophobia (26.9%), and decreased appetite, dry eye, nausea, and thrombocytopenia (23.1% each). No treatment-related grade 4-5 AEs were reported. Systemic exposure of PF-06263507 increased in a dose-related manner. At the maximum tolerated dose (MTD, 4.34 mg/kg), mean terminal half-life for PF-06263507 and unconjugated payload were ~6 and 3 days, respectively. Payload serum concentrations were substantially lower compared with PF-06263507. No objective responses were observed. Conclusions The MTD and recommended phase II dose were determined to be 4.34 mg/kg. Ocular toxicities accounted for the DLTs observed, as previously reported with monomethylauristatin-F payloads. Further studies are warranted to investigate clinical activity of this agent in patients with 5T4-expressing tumors.Trial registration ID: NCT01891669.


Subject(s)
Antibodies, Monoclonal, Humanized , Antigens, Neoplasm , Immunoconjugates , Membrane Glycoproteins/antagonists & inhibitors , Neoplasms/drug therapy , Oligopeptides , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/therapeutic use , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Female , Humans , Immunoconjugates/adverse effects , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Keratitis/chemically induced , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Oligopeptides/adverse effects , Oligopeptides/pharmacokinetics , Oligopeptides/therapeutic use , Photophobia/chemically induced , Treatment Outcome
10.
Immunotherapy ; 8(10): 1175-86, 2016 10.
Article in English | MEDLINE | ID: mdl-27468136

ABSTRACT

AIM: To assess the long-term efficacy, safety and tolerability of recombinant human hyaluronidase-facilitated subcutaneous infusion of immunoglobulin (Ig) (fSCIG; HYQVIA(®); IGHy) in children aged <18 years. PATIENTS & METHODS: Patients with primary immunodeficiency diseases were included in the studies. IGHy was administered every 3 or 4 weeks. RESULTS: Validated acute serious bacterial infections were reported at 0.08/patient-year (four pneumonia episodes in three patients). No serious adverse drug reaction (ADR) was reported, and rates of local and systemic ADRs were low (0.09/infusion and 0.1/infusion). Infection rates were low (3.02/patient-year) with sustained Ig trough levels (median: 1009 mg/dl). Of 674 IGHy infusions, 97.2% required no change of administration due to ADR, in most (82.5%) with one infusion site. No patient developed neutralizing anti-rHuPH20 antibodies. Postpivotal study, 100% of patients aged <14 years or their caregivers and 85.7% of patients aged 14 to <18 years expressed preference for IGHy compared with Ig administered intravenously or Ig administered subcutaneously. CONCLUSION: These studies, with the longest (maximum: 3.3 years) duration of any reported Ig replacement trials in children with primary immunodeficiency diseases, showed low infection, local and systemic reaction rates along with well-tolerated infusions given in a single site.


Subject(s)
Antigens, Neoplasm/therapeutic use , Histone Acetyltransferases/therapeutic use , Hyaluronoglucosaminidase/therapeutic use , Immunoglobulins/therapeutic use , Immunologic Deficiency Syndromes/therapy , Adolescent , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/genetics , Child , Child, Preschool , Female , Histone Acetyltransferases/adverse effects , Histone Acetyltransferases/genetics , Humans , Hyaluronoglucosaminidase/adverse effects , Hyaluronoglucosaminidase/genetics , Immunologic Deficiency Syndromes/immunology , Injections, Subcutaneous , Male , Recombinant Proteins/genetics , Time Factors , United States
11.
Pharmacol Ther ; 165: 32-49, 2016 09.
Article in English | MEDLINE | ID: mdl-27235391

ABSTRACT

DNA vaccination represents a smart and promising approach to cancer immunotherapy. DNA vaccines for cancer immunotherapy are designed to deliver one or several genes encoding tumor antigens, thereby eliciting or augmenting antigen-specific immune responses against antigens that play a central role in tumor initiation, progression and metastasis. Vaccine efficacy can be significantly improved by implementing strategies for enhancing antigen presentation and immunogenicity, such as new delivery systems, addition of molecular adjuvants and immunostimulatory signals, optimized prime-boost strategies or blockade of immune checkpoints. Taken into consideration that innate immune responses are important in the induction and enhancement of antigen-specific adaptive responses, manipulations that integrate these approaches in the vaccine design can achieve activation of protective adaptive immune responses, thereby overcoming the self-tolerance towards many tumor antigens. Such approaches are employed in a number of clinical trials for DNA cancer immunotherapy and hold promise for prophylactic and therapeutic vaccine development. In this context, strategies that improve immunogenicity and enhance the efficacy of DNA vaccines for cancer immunotherapy are discussed.


Subject(s)
Antigens, Neoplasm/therapeutic use , Cancer Vaccines/therapeutic use , Genetic Therapy/methods , Immunotherapy/methods , Neoplasms/therapy , Vaccines, DNA/therapeutic use , Adjuvants, Immunologic/therapeutic use , Animals , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Genetic Therapy/adverse effects , Humans , Immunogenicity, Vaccine , Immunotherapy/adverse effects , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Tumor Escape , Tumor Microenvironment , Vaccines, DNA/adverse effects , Vaccines, DNA/genetics , Vaccines, DNA/immunology
12.
Neuro Oncol ; 18(8): 1157-68, 2016 08.
Article in English | MEDLINE | ID: mdl-26984745

ABSTRACT

BACKGROUND: Low-grade gliomas (LGGs) are the most common brain tumors of childhood. Although surgical resection is curative for well-circumscribed superficial lesions, tumors that are infiltrative or arise from deep structures are therapeutically challenging, and new treatment approaches are needed. Having identified a panel of glioma-associated antigens (GAAs) overexpressed in these tumors, we initiated a pilot trial of vaccinations with peptides for GAA epitopes in human leukocyte antigen-A2+ children with recurrent LGG that had progressed after at least 2 prior regimens. METHODS: Peptide epitopes for 3 GAAs (EphA2, IL-13Rα2, and survivin) were emulsified in Montanide-ISA-51 and administered subcutaneously adjacent to intramuscular injections of polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose every 3 weeks for 8 courses, followed by booster vaccines every 6 weeks. Primary endpoints were safety and T-lymphocyte responses against GAA epitopes. Treatment response was evaluated clinically and by MRI. RESULTS: Fourteen children were enrolled. Other than grade 3 urticaria in one child, no regimen-limiting toxicity was encountered. Vaccination induced immunoreactivity to at least one vaccine-targeted GAA in all 12 evaluable patients: to IL-13Rα2 in 3, EphA2 in 11, and survivin in 3. One child with a metastatic LGG had asymptomatic pseudoprogression noted 6 weeks after starting vaccination, followed by dramatic disease regression with >75% shrinkage of primary tumor and regression of metastatic disease, persisting >57 months. Three other children had sustained partial responses, lasting >10, >31, and >45 months, and one had a transient response. CONCLUSIONS: GAA peptide vaccination in children with recurrent LGGs is generally well tolerated, with preliminary evidence of immunological and clinical activity.


Subject(s)
Antigens, Neoplasm/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Carboxymethylcellulose Sodium/analogs & derivatives , Glioma/drug therapy , Glioma/immunology , Interferon Inducers/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Vaccination/methods , Adolescent , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/immunology , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/adverse effects , Carboxymethylcellulose Sodium/therapeutic use , Child , Child, Preschool , Disease-Free Survival , Epitopes , Female , Humans , Infant , Inhibitor of Apoptosis Proteins/immunology , Interferon Inducers/administration & dosage , Interferon Inducers/adverse effects , Interferon Inducers/immunology , Interleukin-13 Receptor alpha2 Subunit/immunology , Male , Neoplasm Grading , Pilot Projects , Poly I-C/administration & dosage , Poly I-C/adverse effects , Poly I-C/immunology , Polylysine/administration & dosage , Polylysine/adverse effects , Polylysine/immunology , Polylysine/therapeutic use , Receptor, EphA2/immunology , Survivin , Treatment Outcome
13.
PLoS One ; 11(3): e0150733, 2016.
Article in English | MEDLINE | ID: mdl-26983031

ABSTRACT

OBJECTIVE: No efficient medical treatment is available for severe acute hepatitis (SAH) except N-acetylcysteine for acetaminophen-induced acute liver failure. The human C-type lectin Reg3α, referred to as ALF-5755, improved survival in an animal model of acute liver failure and was well tolerated in a phase 1 trial in humans. We performed a phase 2a trial of ALF5755 in non-acetaminophen induced SAH. DESIGN: double-blind, randomized, placebo-controlled study. The primary end-point was the improvement in the coagulation protein synthesis assessed by the change of Prothrombin (PR) during the 72 hours following treatment initiation calculated as PRH0 minus PRH72 divided by 72 (PR slope H0H72). Intention to treat (ITT) and per-protocol (PP) analysis of the entire group and the Hepatitis B virus (HBV)/AIH (auto-immune hepatitis) sub-group were done separately. RESULTS: 57 patients were included. Twenty-eight received ALF-5755, 29 the placebo. Etiologies were: Hepatitis A (n = 10), HBV (n = 13), AIH (n = 9), drug-induced (n = 8), other (n = 17). On the whole group, nor the PR slope H0H72 (0.18±0.31 vs 0.25±0.32), nor the transplant-free survival rate at day 21 (75 vs 86%) differed between groups. Conversely, in the HBV-AIH subgroup, in which ALF was more severe, PR slope H0-H72 was higher in the ALF-5755 arm, the difference being significant in PP analysis (0.048±0.066 vs -0.040±0.099, p = 0.04); the median length of hospitalization was lower in the ALF-5755 group (8 vs 14 days, p = 0.02). CONCLUSION: ALF-5755 was not efficient in a ITT analysis performed on the whole sample; however it led to a significant, although moderate, clinical benefit in a PP analysis of the sub-group of patients with HBV or AIH related SAH. As HBV is the major cause of SAH in Asia and Africa and AIH a growing cause, this study emphasizes the need to pursuit the evaluation of this novel medical treatment of SAH. TRIAL REGISTRATION: ClinicalTrials.gov NCT01318525.


Subject(s)
Antigens, Neoplasm/therapeutic use , Antioxidants/therapeutic use , Biomarkers, Tumor/therapeutic use , Extracellular Matrix/drug effects , Lectins, C-Type/therapeutic use , Liver Diseases/drug therapy , Recombinant Proteins/therapeutic use , Acute Disease , Adult , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/pharmacology , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Area Under Curve , Biomarkers, Tumor/adverse effects , Biomarkers, Tumor/pharmacokinetics , Biomarkers, Tumor/pharmacology , Double-Blind Method , Female , Humans , Male , Middle Aged , Pancreatitis-Associated Proteins , Placebos , Prognosis , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
14.
J Appl Toxicol ; 35(7): 717-28, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25219328

ABSTRACT

The MAGE-A3 recombinant protein combined with AS15 immunostimulant (MAGE-A3 Cancer Immunotherapeutic) is under development by GlaxoSmithKline for the treatment of lung cancer and melanoma. We performed non-clinical safety studies evaluating potential local and systemic toxic effects induced by MAGE-A3 Cancer Immunotherapeutic in rabbits (study 1) and cynomolgus monkeys (study 2). Animals were allocated to two groups to receive a single (rabbits) or 25 repeated (every 2 weeks) injections (monkeys) of MAGE-A3 Cancer Immunotherapeutic (treatment groups) or saline (control groups). All rabbits were sacrificed 3 days post-injection and monkeys 3 days following last injection (3/5 per gender per group) or after a 3-month treatment-free period (2/5 per gender per group). Local and systemic reactions and MAGE-A3-specific immune responses (monkeys) were assessed. Macroscopic and microscopic (for rabbits, injection site only) post-mortem examinations were performed on all animals. No systemic toxicity or unscheduled mortalities were recorded. Single (rabbits) and repeated (monkeys; up to four times at the same site) injections were well tolerated. Following five to seven repeated injections, limb circumferences increased up to 26% (5 h post-injection), but returned to normal after 1-8 days. Three days after the last injection, enlargements of iliac, popliteal, axillary and inguinal lymph nodes, and increased incidence or severity of mononuclear inflammatory cell infiltrates was observed in injected muscles of treated monkeys. No treatment-related macroscopic findings were recorded after the treatment-free period. MAGE-A3-specific antibody and T-cell responses were raised in all treated monkeys, confirming test item exposure. Single or repeated intramuscular injections of MAGE-A3 Cancer Immunotherapeutic were well tolerated in rabbits and monkeys.


Subject(s)
Antigens, Neoplasm/adverse effects , Neoplasm Proteins/adverse effects , Animals , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/therapeutic use , Drug Administration Schedule , Female , Immunotherapy/methods , Injections, Intramuscular , Macaca fascicularis , Male , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/therapeutic use , Neoplasms/drug therapy , Rabbits , T-Lymphocytes/drug effects , T-Lymphocytes/physiology
15.
Thyroid ; 24(2): 390-4, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23590991

ABSTRACT

BACKGROUND: Immunotherapies and targeted therapies are frequently associated with thyroid dysfunction, which is in contrast with the rare thyroid abnormalities induced by cytotoxic agents. Immunotherapy with NY-ESO-1, a tumor-associated antigen expressed by a number of malignancies, was reported to trigger hyperthyroidism or hypothyroidism in two HLA-A2 patients with ovarian cancer. We describe now a case of Graves' disease triggered by NY-ESO-1 in a HLA-A2-negative woman. PATIENT FINDINGS: A 32-year-old woman with a synovial sarcoma received radiotherapy, chemotherapy, and finally NY-ESO-1 vaccine. The patient was found to have HLA A11/A33(19), B13/B56(22), Cw3/-. One month after the beginning of immunotherapy, thyroid dysfunction was clinically suspected and Graves' disease was biochemically confirmed. Fearful of the antithyroid drugs' side effects, the patient was treated with a beta-blocker (propranolol, 80-20 mg/day). As hyperthyroidism progressively worsened, the patient underwent total thyroidectomy. We hypothesized that NY-ESO-1 shared partial homology with thyroid autoantigens (the so-called molecular mimicry mechanism) and that at least one pair of homologous sequences contained amino acid sequence binding motifs to a restricted number of HLA molecules. We used BLAST software to search amino acid sequence homologies between NY-ESO-1 and thyroid autoantigens (thyrotropin receptor [TSH-R], thyroperoxidase, and thyroglobulin), and the HLA ligand/motif database to look for HLA/T-cell receptor binding motifs in the regions of NY-ESO-1 and thyroid autoantigens that were homologous. We found 15 epitopic regions of NY-ESO-1 homologous to 15 regions of thyroid autoantigens, some of which epitopic: 5 of TSH-R, 8 of thyroglobulin, and 2 of thyroperoxidase. These homologous sequences contain binding motifs belonging to several HLA class I antigens, including HLA A2 and the patient's A11 and A33. SUMMARY: Genetically predisposed patients who receive NY-ESO-1 vaccination are at risk to develop thyroid dysfunction. CONCLUSIONS: Considering the increasing use of NY-ESO-1, thyroid dysfunctions induced by NY-ESO-1 are expected to increase in cancer patients over the next years.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/adverse effects , Graves Disease/chemically induced , Membrane Proteins/immunology , Sarcoma, Synovial/therapy , Adult , Antigens, Neoplasm/adverse effects , Combined Modality Therapy , Epitopes , Female , Humans , Membrane Proteins/adverse effects , Sequence Homology, Amino Acid , Vaccination/adverse effects
16.
Clin Ther ; 34(11): 2232-45, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23062548

ABSTRACT

BACKGROUND: Alternative treatment of dehydration is needed when intravenous (IV) or oral rehydration therapy fails. Subcutaneous (SC) hydration facilitated by recombinant human hyaluronidase offers an alternative treatment for dehydration. This clinical trial is the first to compare recombinant human hyaluronidase-facilitated SC (rHFSC) rehydration with standard IV rehydration for use in dehydrated children. OBJECTIVE: This Phase IV noninferiority trial evaluated whether rHFSC fluid administration can be given safely and effectively, with volumes similar to those delivered intravenously, to children who have mild to moderate dehydration. METHODS: The study included mild to moderately dehydrated children (Gorelick dehydration score) aged 1 month to 10 years. They were randomized to receive 20 mL/kg of isotonic fluids using rHFSC or IV therapy over 1 hour and then as needed until clinically rehydrated. The primary outcome was total volume of fluid administered (emergency department [ED] plus inpatient hospitalization). Secondary outcomes included mean volume infused in the ED alone, postinfusion dehydration scores and weight changes, line placement success and time, safety, and provider and parent/guardian questionnaire. RESULTS: 148 patients (mean age, 2.3 [1.91] years]; white, 53.4%; black, 31.8%) were enrolled in the intention-to-treat population (73 rHFSC; 75 IV). The primary outcome, mean total volume infused, was 365.0 (324.6) mL in the rHFSC group over 3.1 hours versus 455.8 (597.4) mL in the IV group over 6.6 hours (P = 0.51). The secondary outcome of mean volume infused in the ED alone was 334.3 (226.40) mL in the rHFSC group versus 299.6 (252.33) mL in the IV group (P = 0.03). Dehydration scores and weight changes postinfusion were similar. Successful line placement occurred in all 73 rHFSC-treated patients and 59 of 75 (78.7%) IV-treated patients (P < 0.0001). All IV failures occurred in patients aged <3 years; rHFSC rescue was successful in all patients in whom it was attempted. Both treatments were well tolerated. Clinicians rated fluid administration as easy to perform in 94.5% (69 of 73) of the rHFSC group versus 65.3% (49 of 75) of the IV group (P < 0.001). Parents/caregivers were satisfied or very satisfied with fluid administration in 94.5% (69 of 73) of rHFSC-treated patients and 73.3% (55 of 75) of IV-treated patients. CONCLUSIONS: In mild to moderately dehydrated children, rHFSC was inferior to IV hydration for the primary outcome measure. However, rHFSC was noninferior in the ED phase of hydration. Additional benefits of rHFSC included time and success of line placement, ease of use, and satisfaction. SC hydration facilitated with recombinant human hyaluronidase represents a reasonable addition to the treatment options for children who have mild to moderate dehydration, especially those with difficult IV access. ClinicalTrials.gov identifier: NCT00773175.


Subject(s)
Antigens, Neoplasm/administration & dosage , Dehydration/therapy , Emergency Service, Hospital , Fluid Therapy/methods , Histone Acetyltransferases/administration & dosage , Hyaluronoglucosaminidase/administration & dosage , Hypodermoclysis , Isotonic Solutions/administration & dosage , Antigens, Neoplasm/adverse effects , Body Weight , Child , Child, Preschool , Dehydration/diagnosis , Female , Fluid Therapy/adverse effects , Histone Acetyltransferases/adverse effects , Hospitalization , Humans , Hyaluronoglucosaminidase/adverse effects , Hypodermoclysis/adverse effects , Infant , Infusions, Intravenous , Isotonic Solutions/adverse effects , Male , Prospective Studies , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Severity of Illness Index , Surveys and Questionnaires , Time Factors , Treatment Outcome , United States
17.
J Diabetes Sci Technol ; 6(4): 764-72, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22920800

ABSTRACT

Rapid-acting insulin analogs were introduced to the market in the 1990s, and these products have improved treatment of diabetes by shortening the optimum delay time between injections and meals. Compared with regular human insulin, rapid-acting insulin formulations also reduce postprandial glycemic excursions while decreasing risk of hypoglycemia. However, the current prandial products are not fast enough for optimum convenience or control. Recombinant human hyaluronidase (rHuPH20) has been used to increase the dispersion and absorption of other injected drugs, and in the case of prandial insulin analogs, it confers both ultrafast absorption and action profiles. Animal toxicology studies have demonstrated excellent tolerability of rHuPH20, and human studies, involving over 60,000 injections of prandial insulin + rHuPH20 to date, have similarly shown excellent safety and tolerability. Studies using rapid-acting analog insulin with rHuPH20 have included clinic-based pharmacokinetic and glucodynamic euglycemic glucose clamp studies, test meal studies, and take-home treatment studies. Administration methods have included subcutaneous injection of coformulations of rapid-acting insulin + rHuPH20 as well as continuous subcutaneous infusion of coformulations or use of pretreatment of newly inserted infusion sets with rHuPH20 followed by standard continuous subcutaneous insulin infusion therapy. These studies have demonstrated acceleration of insulin absorption and action along with improvement in postprandial glycemic excursions and reduction in hypoglycemia risks. Further, rHuPH20 reduces intrasubject variability of insulin absorption and action and provides greater consistency in absorption and action profiles over wear time of an infusion set. Further studies of rHuPH20 in the take-home treatment setting are underway.


Subject(s)
Antigens, Neoplasm/pharmacology , Chemistry, Pharmaceutical/methods , Histone Acetyltransferases/pharmacology , Hyaluronoglucosaminidase/pharmacology , Insulin, Short-Acting/pharmacokinetics , Absorption/drug effects , Acceleration , Adult , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/chemistry , Dosage Forms , Excipients/administration & dosage , Excipients/adverse effects , Excipients/chemistry , Excipients/pharmacology , Histone Acetyltransferases/administration & dosage , Histone Acetyltransferases/adverse effects , Histone Acetyltransferases/chemistry , Humans , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/adverse effects , Hyaluronoglucosaminidase/chemistry , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Infusions, Subcutaneous , Injections, Subcutaneous , Insulin Infusion Systems , Insulin Lispro/administration & dosage , Insulin Lispro/adverse effects , Insulin Lispro/pharmacokinetics , Insulin, Short-Acting/administration & dosage , Insulin, Short-Acting/adverse effects , Insulin, Short-Acting/chemistry , Male , Postprandial Period/drug effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
19.
Endocr Pract ; 17(6): 914-21, 2011.
Article in English | MEDLINE | ID: mdl-22138081

ABSTRACT

OBJECTIVE: To discuss clinical studies in which recombinant human hyaluronidase (rHuPH20) was used to increase insulin dispersion and accelerate its absorption. METHODS: We reviewed 10 pertinent clinical studies, 8 of which had data available. RESULTS: In 4 euglycemic clamp studies, coinjection of rHuPH20 consistently yielded acceleration of insulin absorption, providing twice the insulin exposure during the first hour, greater and earlier peak exposure, and half the exposure beyond 2 hours after injection. Insulin-action profiles were similarly accelerated, with a 15-minute faster onset of insulin action and a 45-minute shorter duration of action for each of the 3 commercial rapid-acting insulin analogues. Infusion aspart insulin formulated with rHuPH20 also accelerated insulin absorption and action over the infusion set life when delivered by insulin pump. Administration of rHuPH20 reduced the inconsistency of insulin absorption and action profiles attributable to 3 factors-lack of reproducibility after identical injections, differences across insulin dose ranges, and changes over infusion site life. The rHuPH20-facilitated ultrafast profile consistently reduced hyperglycemic excursions both in injections immediately preceding liquid test meals and in bolus infusions immediately before solid test meals. rHuPH20-facilitated insulin administration has been well tolerated, with safety and tolerability similar to those with the comparator insulin alone. CONCLUSION: rHuPH20 accelerates insulin-action profiles to an extent comparable to the difference between rapid-acting insulin analogue profiles and those of regular insulin. Studies are currently under way to characterize the effect on diabetes management end points (including hemoglobin A1c, blood glucose, and rates of hyperglycemia) of insulin analogues coformulated with rHuPH20 for treatment of both type 1 and type 2 diabetes.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Antigens, Neoplasm/pharmacology , Diabetes Mellitus/drug therapy , Histone Acetyltransferases/pharmacology , Hyaluronoglucosaminidase/pharmacology , Hyperglycemia/prevention & control , Hypoglycemic Agents/pharmacokinetics , Insulin Aspart/pharmacokinetics , Absorption/drug effects , Adjuvants, Pharmaceutic/administration & dosage , Adjuvants, Pharmaceutic/adverse effects , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/adverse effects , Diabetes Mellitus/blood , Drug Combinations , Drug Synergism , Histone Acetyltransferases/administration & dosage , Histone Acetyltransferases/adverse effects , Humans , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/adverse effects , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Hypoglycemic Agents/therapeutic use , Injections, Subcutaneous , Insulin Aspart/administration & dosage , Insulin Aspart/blood , Insulin Aspart/therapeutic use , Insulin Infusion Systems , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology
20.
Clin Cancer Res ; 17(4): 861-70, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21163871

ABSTRACT

PURPOSE: NY-ESO-1, one of the most immunogenic tumor antigens, is expressed in 15% to 25% of metastatic prostate cancers. The immunological and clinical effects of vaccination with recombinant NY-ESO-1 protein combined with CpG as adjuvant were evaluated. EXPERIMENTAL DESIGN: In a phase I clinical study, patients with advanced prostate cancer were vaccinated with recombinant NY-ESO-1 protein (100 µg) mixed with CpG 7909 (2.5 mg) every 3 weeks intradermally for 4 doses. Objectives of the study were the safety of the vaccine and changes of specific humoral and cellular immunological responses to NY-ESO-1 in relation to detectable NY-ESO-1 expression in the individual tumor. RESULTS: All 12 baseline sero-negative patients developed high-titer NY-ESO-1 antibody responses. B-cell epitope mapping identified NY-ESO-1 p91-110 to be recognized most frequently by vaccine-induced antibodies. Two patients developed significant antibody titers against the adjuvant CpG. NY-ESO-1-specific CD4+ and/or CD8+ T-cell responses were induced in 9 patients (69%). Five of these 9 patients did not express NY-ESO-1 in the autologous tumor. Postvaccine CD8+ T-cell clones recognized and lyzed HLA-matched tumor cell lines in an antigen-specific manner. CONCLUSION: Our data provide clear evidence for the capacity of NY-ESO-1 protein/CpG vaccine to induce integrated antigen-specific immune responses in vivo and to efficiently prime CD8+ T-cell responses in NY-ESO-1 antigen-negative patients. Our results may also support further clinical vaccination protocols with NY-ESO-1 protein not only focused on the treatment of existing cancer, but also to prevent further development of NY-ESO-1 positive cancers in vivo.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Membrane Proteins/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Prostatic Neoplasms/therapy , Adaptive Immunity , Adjuvants, Immunologic/adverse effects , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/adverse effects , Chemotherapy, Adjuvant , Humans , Injections, Intradermal , Male , Membrane Proteins/adverse effects , Membrane Proteins/immunology , Oligodeoxyribonucleotides/adverse effects , Oligodeoxyribonucleotides/immunology , Prostatic Neoplasms/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...