Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 62
1.
Cell Rep Med ; 2(10): 100422, 2021 10 19.
Article En | MEDLINE | ID: mdl-34755134

Provoked by sterile/nonsterile insults, prolonged monocyte mobilization and uncontrolled monocyte/macrophage activation can pose imminent or impending harm to the affected organs. Curiously, folate receptor beta (FRß), with subnanomolar affinity for the vitamin folic acid (FA), is upregulated during immune activation in hematopoietic cells of the myeloid lineage. This phenomenon has inspired a strong interest in exploring FRß-directed diagnostics/therapeutics. Previously, we have reported that FA-targeted aminopterin (AMT) therapy can modulate macrophage function and effectively treat animal models of inflammation. Our current investigation of a lead compound (EC2319) leads to discovery of a highly FR-specific mechanism of action independent of the root causes against inflammatory monocytes. We further show that EC2319 suppresses interleukin-6/interleukin-1ß release by FRß+ monocytes in a triple co-culture leukemic model of cytokine release syndrome with anti-CD19 chimeric antigen receptor T cells. Because of its chemical stability and metabolically activated linker, EC2319 demonstrates favorable pharmacokinetic characteristics and cross-species translatability to support future pre-clinical and clinical development.


Aminopterin/pharmacology , Cytokine Release Syndrome/prevention & control , Folate Receptor 2/genetics , Folic Acid Antagonists/pharmacology , Folic Acid/metabolism , Macrophages/drug effects , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , CHO Cells , Cricetulus , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Female , Folate Receptor 1/antagonists & inhibitors , Folate Receptor 1/genetics , Folate Receptor 1/immunology , Folate Receptor 2/antagonists & inhibitors , Folate Receptor 2/immunology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Models, Biological , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , RAW 264.7 Cells , Rats , Rats, Inbred Lew , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
2.
Cells ; 10(8)2021 08 03.
Article En | MEDLINE | ID: mdl-34440744

Folic acid has been identified to be integral in rapid tissue growth and cell division during fetal development. Different studies indicate folic acid's importance in improving childhood behavioral outcomes and underline its role as a modifiable risk factor for autism spectrum disorders. The aim of this systematic review is to both elucidate the potential role of folic acid in autism spectrum disorders and to investigate the mechanisms involved. Studies have pointed out a potential beneficial effect of prenatal folic acid maternal supplementation (600 µg) on the risk of autism spectrum disorder onset, but opposite results have been reported as well. Folic acid and/or folinic acid supplementation in autism spectrum disorder diagnosed children has led to improvements, both in some neurologic and behavioral symptoms and in the concentration of one-carbon metabolites. Several authors report an increased frequency of serum auto-antibodies against folate receptor alpha (FRAA) in autism spectrum disorder children. Furthermore, methylene tetrahydrofolate reductase (MTHFR) polymorphisms showed a significant influence on ASD risk. More clinical trials, with a clear study design, with larger sample sizes and longer observation periods are necessary to be carried out to better evaluate the potential protective role of folic acid in autism spectrum disorder risk.


Autism Spectrum Disorder/pathology , Folic Acid/metabolism , Autism Spectrum Disorder/metabolism , Autoantibodies/blood , Dietary Supplements , Folate Receptor 1/immunology , Folic Acid/administration & dosage , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Single Nucleotide , Risk Factors
3.
Anticancer Res ; 41(4): 1811-1819, 2021 Apr.
Article En | MEDLINE | ID: mdl-33813386

BACKGROUND/AIM: Glioblastoma is the most common cancer among primary brain tumors, however, its prognosis and treatment advances are very poor. Here, we investigated whether c-Met, FOLR1, and AXL proteins are promising targets for chimeric antigen receptor (CAR) T-cell therapy, for they are known to be over-expressed in a variety of solid tumors. MATERIALS AND METHODS: CAR constructs were prepared and CAR KHYG-1 cells targeting c-Met, FOLR1, or AXL were made by lentiviral transduction. The activity of CAR KHYG-1 cells against cancer cells was measured by cytokine secretion and cell lysis assays. RESULTS: c-Met and AXL were over-expressed in most glioblastoma cell lines (11/13), but not in neuroblastoma cell lines (0/8). FOLR1 was over-expressed only in one among 16 glioblastoma cell lines. Our antigen-specific CAR KHYG-1 cells eradicated target positive glioblastoma cells selectively. CONCLUSION: Anti-c-Met and anti-AXL CAR NK or T cells could be effective in glioblastoma cells.


Brain Neoplasms/therapy , Glioblastoma/therapy , Immunotherapy, Adoptive , Killer Cells, Natural/immunology , Proto-Oncogene Proteins c-met/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Coculture Techniques , Cytokines/metabolism , Cytotoxicity, Immunologic , Folate Receptor 1/immunology , Folate Receptor 1/metabolism , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Jurkat Cells , Killer Cells, Natural/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Axl Receptor Tyrosine Kinase
4.
Gynecol Oncol ; 160(2): 520-529, 2021 02.
Article En | MEDLINE | ID: mdl-33342620

OBJECTIVE: Chimeric antigen receptor (CAR)-T cell strategies ideally target a surface antigen that is exclusively and uniformly expressed by tumors; however, no such antigen is known for high-grade serous ovarian carcinoma (HGSC). A potential solution involves combinatorial antigen targeting with AND or OR logic-gating. Therefore, we investigated co-expression of CA125, Mesothelin (MSLN) and Folate Receptor alpha (FOLRA) on individual tumor cells in HGSC. METHODS: RNA expression of CA125, MSLN, and FOLR1 was assessed using TCGA (HGSC) and GTEx (healthy tissues) databases. Antigen expression profiles and CD3+, CD8+ and CD20+ tumor-infiltrating lymphocyte (TIL) patterns were assessed in primary and recurrent HGSC by multiplex immunofluorescence and immunohistochemistry. RESULTS: At the transcriptional level, each antigen was overexpressed in >90% of cases; however, MSLN and FOLR1 showed substantial expression in healthy tissues. At the protein level, CA125 was expressed by the highest proportion of cases and tumor cells per case, followed by MSLN and FOLRA. The most promising pairwise combination was CA125 and/or MSLN (OR gate), with 51.9% of cases containing ≥90% of tumor cells expressing one or both antigens. In contrast, only 5.8% of cases contained ≥90% of tumor cells co-expressing CA125 and MSLN (AND gate). Antigen expression patterns showed modest correlations with TIL. Recurrent tumors retained expression of all three antigens and showed increased TIL densities. CONCLUSIONS: An OR-gated CAR-T cell strategy against CA125 and MSLN would target the majority of tumor cells in most cases. Antigen expression and T-cell infiltration patterns are favorable for this strategy in primary and recurrent disease.


Antigens, Neoplasm/metabolism , Carcinoma, Ovarian Epithelial/immunology , Immunotherapy, Adoptive/methods , Neoplasm Recurrence, Local/immunology , Ovarian Neoplasms/immunology , Receptors, Chimeric Antigen/metabolism , Antigens, Neoplasm/immunology , CA-125 Antigen/immunology , CA-125 Antigen/metabolism , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/therapy , Female , Folate Receptor 1/immunology , Folate Receptor 1/metabolism , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Gene Expression Profiling , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mesothelin , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Ovary/immunology , Ovary/pathology , Receptors, Chimeric Antigen/immunology
5.
Nat Commun ; 11(1): 5173, 2020 10 14.
Article En | MEDLINE | ID: mdl-33057068

In ovarian cancer (OC), IL-17-producing T cells (Th17s) predict improved survival, whereas regulatory T cells predict poorer survival. We previously developed a vaccine whereby patient-derived dendritic cells (DCs) are programmed to induce Th17 responses to the OC antigen folate receptor alpha (FRα). Here we report the results of a single-arm open-label phase I clinical trial designed to determine vaccine safety and tolerability (primary outcomes) and recurrence-free survival (secondary outcome). Immunogenicity is also evaluated. Recruitment is complete with a total of 19 Stage IIIC-IV OC patients in first remission after conventional therapy. DCs are generated using our Th17-inducing protocol and are pulsed with HLA class II epitopes from FRα. Mature antigen-loaded DCs are injected intradermally. All patients have completed study-related interventions. No grade 3 or higher adverse events are seen. Vaccination results in the development of Th1, Th17, and antibody responses to FRα in the majority of patients. Th1 and antibody responses are associated with prolonged recurrence-free survival. Antibody-dependent cell-mediated cytotoxic activity against FRα is also associated with prolonged RFS. Of 18 patients evaluable for efficacy, 39% (7/18) remain recurrence-free at the time of data censoring, with a median follow-up of 49.2 months. Thus, vaccination with Th17-inducing FRα-loaded DCs is safe, induces antigen-specific immunity, and is associated with prolonged remission.


Cancer Vaccines/administration & dosage , Dendritic Cells/transplantation , Neoplasm Recurrence, Local/epidemiology , Ovarian Neoplasms/therapy , Th17 Cells/immunology , Aged , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Disease-Free Survival , Female , Folate Receptor 1/immunology , Humans , Immunity, Humoral , Injections, Intradermal , Interferon-gamma/metabolism , Interleukin-17/metabolism , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Ovarian Neoplasms/immunology , Ovarian Neoplasms/mortality , Th17 Cells/metabolism , Transplantation, Autologous/adverse effects , Transplantation, Autologous/methods
6.
Semin Pediatr Neurol ; 35: 100835, 2020 10.
Article En | MEDLINE | ID: mdl-32892962

Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder that currently has no approved medical therapy to address core symptoms or underling pathophysiological processes. Several compounds are under development that address both underlying pathophysiological abnormalities and core ASD symptoms. This article reviews one of these treatments, d,l-leucovorin calcium (also known as folinic acid) for treatment of folate pathway abnormalities in children with ASD. Folate is a water-soluble B vitamin that is essential for normal neurodevelopment and abnormalities in the folate and related pathways have been identified in children with ASD. One of these abnormalities involves a partial blockage in the ability of folate to be transported into the brain utilizing the primary transport mechanism, the folate receptor alpha. Autoantibodies which interfere with the function of the folate receptor alpha called folate receptor alpha autoantibodies have been identified in 58%-76% of children with ASD and independent studies have demonstrated that blood titers of these autoantibodies correlate with folate levels in the cerebrospinal fluid. Most significantly, case-series, open-label, and single and double-blind placebo-controlled studies suggest that d,l-leucovorin, a reduced folate that can bypass the blockage at the folate receptor alpha by using the reduced folate carrier, an alternate pathway, can substantially improve particular symptoms in children with ASD, especially those positive for folate receptor alpha autoantibodies. This article reviews the current evidence for treating core and associated symptoms and underlying pathophysiological mechanisms in children with ASD with d,l-leucovorin.


Autism Spectrum Disorder , Folate Receptor 1/immunology , Folic Acid Deficiency , Folic Acid , Leucovorin/pharmacology , Vitamin B Complex/pharmacology , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/immunology , Autism Spectrum Disorder/metabolism , Autoantibodies , Child , Folic Acid/cerebrospinal fluid , Folic Acid Deficiency/cerebrospinal fluid , Folic Acid Deficiency/drug therapy , Humans , Leucovorin/administration & dosage , Vitamin B Complex/administration & dosage
7.
Ann Diagn Pathol ; 48: 151596, 2020 Oct.
Article En | MEDLINE | ID: mdl-32829070

Folate receptor alpha (FRα) is a membrane-bound protein with a high affinity for folate, which is necessary for the biosynthesis of amino acids and nucleotide bases. It has been shown to be a potential prognostic and therapeutic target, primarily in lung and ovarian cancer, as well as in breast cancer. The aim of this study was to examine FRα expression in a cohort of patients with triple negative breast cancer (TNBC), in correlation with clinicopathological parameters and prognostic factors. By using polyclonal FRα antibody on archival paraffin blocks immunohistochemistry was performed. To evaluate the expression of FRα, H-score was used, which marks both the proportion of stained cells and the intensity of staining. Statistical analysis correlating FRα expression with clinicopathologic parameters and clinical outcome were performed. FRα was expressed in most of the patients (85%). Significant correlation of expression and histologic grade (Mann Whitney U test, P = 0,03) and type of tumor (P = 0,02), was found. It was noticed that with higher Ki-67 proliferation index values, H-score has lower values (r = -0,284, P = 0,006). Multivariant regression analysis (Cox regression, Stepwise method) showed H-score as a significant predictor for the risk of disease recurrence (OR = 1,005, P = 0,04). No correlation between FRα expression and overall survival (OS) and disease-free survival (DFS) was found. In conclusion, FRα is highly expressed in TNBC, and, given the correlation with clinicopathological parameters, subpopulation of patients could be identified that could be potential targets for new therapeutic perspectives in the treatment of this breast cancer subtype.


Folate Receptor 1/biosynthesis , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma/diagnosis , Carcinoma/epidemiology , Disease-Free Survival , Female , Folate Receptor 1/immunology , Folate Receptor 1/metabolism , Humans , Immunohistochemistry/methods , Middle Aged , Neoplasm Grading/methods , Neoplasm Recurrence, Local/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phenotype , Prognosis , Research Design/statistics & numerical data , Retrospective Studies , Risk Assessment , Triple Negative Breast Neoplasms/mortality
8.
Cancer Lett ; 491: 121-131, 2020 10 28.
Article En | MEDLINE | ID: mdl-32795486

Triple-negative breast cancer (TNBC) is the most complex and challenging breast cancer subtype to treat, and chemotherapy remains the standard of care. Clinically, TNBC has a relatively high rate of recurrence and poor prognosis, which leads to a significant effort to discover novel strategies to treat patients with these tumors. Currently, chimeric antigen receptor (CAR) T cell-based immunotherapy redirects the patient's immune system directly to recognize and eradicate tumor-associated antigens (TAAs) expressing tumor cells being explored as a treatment for TNBC. A steadily increasing research in CAR T-cell therapy targeting different TAAs in TNBC has reported. In this review, we introduce the CAR technology and summarize the potential TAAs, available CARs, the antitumor activity, and the related toxicity of CARs currently under investigation for TNBC. We also highlight the potential strategies to prevent/reduce potential "on target, off tumor" toxicity induced by CAR T-cell therapy. This review will help to explore proper targets to expand further the CAR T-cell therapy for TNBCs in the clinic.


Immunotherapy, Adoptive/methods , Triple Negative Breast Neoplasms/therapy , Antigens, Neoplasm/immunology , Chondroitin Sulfate Proteoglycans/immunology , Female , Folate Receptor 1/immunology , GPI-Linked Proteins/immunology , Humans , Immunotherapy, Adoptive/adverse effects , Intercellular Adhesion Molecule-1/immunology , Membrane Proteins/immunology , Mesothelin , Mucin-1/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptor Tyrosine Kinase-like Orphan Receptors/immunology
9.
J Immunother Cancer ; 8(1)2020 06.
Article En | MEDLINE | ID: mdl-32503949

BACKGROUND: Immune checkpoint inhibitors (ICIs) to date have demonstrated limited activity in advanced ovarian cancer (OC). Folate receptor alpha (FRα) is overexpressed in the majority of OCs and presents an attractive target for a combination immunotherapy to potentially overcome resistance to ICI in OCs. The current study sought to examine clinical and immunologic responses to TPIV200, a multiepitope FRα vaccine administered with programmed death ligand 1 (PD-L1) inhibitor durvalumab in patients with advanced platinum-resistant OC. METHODS: Following Simon two-stage phase II trial design, 27 patients were enrolled. Treatment was administered in 28-day cycles (intradermal TPIV200 and granulocyte-macrophage colony-stimulating factor (GM-CSF) for 6 cycles and intravenous durvalumab for 12 cycles). Primary endpoints included overall response rate and progression-free survival at 24 weeks. Translational parameters focused on tumor microenvironment, PD-L1 and FRα expression, and peripheral vaccine-specific immune responses. RESULTS: Treatment was well tolerated, with related grade 3 toxicity rate of 18.5%. Increased T cell responses to the majority of peptides were observed in all patients at 6 weeks (p<0.0001). There was one unconfirmed partial response (3.7%) and nine patients had stable disease (33.3%). Clinical benefit was not associated with baseline FRα or PD-L1 expression. One patient with prolonged clinical benefit demonstrated loss of FRα expression and upregulation of PD-L1 in a progressing lesion. Despite the low overall response rate, the median overall survival was 21 months (13.5-∞), with evidence of benefit from postimmunotherapy regimens. CONCLUSIONS: Combination of TPIV200 and durvalumab was safe and elicited robust FRα-specific T cell responses in all patients. Unexpectedly durable survival in this heavily pretreated population highlights the need to investigate the impact of FRα vaccination on the OC biology post-treatment.


Antibodies, Monoclonal/therapeutic use , Biomarkers, Tumor/immunology , Cancer Vaccines/therapeutic use , Folate Receptor 1/immunology , Ovarian Neoplasms/drug therapy , Tumor Microenvironment/immunology , Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/immunology , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Antineoplastic Agents, Immunological/therapeutic use , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/pathology , Drug Therapy, Combination , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Non-Randomized Controlled Trials as Topic , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Prognosis , Survival Rate , Treatment Outcome
10.
Nat Commun ; 11(1): 3196, 2020 06 24.
Article En | MEDLINE | ID: mdl-32581215

T-cell bispecific antibodies (TCBs) crosslink tumor and T-cells to induce tumor cell killing. While TCBs are very potent, on-target off-tumor toxicity remains a challenge when selecting targets. Here, we describe a protease-activated anti-folate receptor 1 TCB (Prot-FOLR1-TCB) equipped with an anti-idiotypic anti-CD3 mask connected to the anti-CD3 Fab through a tumor protease-cleavable linker. The potency of this Prot- FOLR1-TCB is recovered following protease-cleavage of the linker releasing the anti-idiotypic anti-CD3 scFv. In vivo, the Prot-FOLR1-TCB mediates antitumor efficacy comparable to the parental FOLR1-TCB whereas a noncleavable control Prot-FOLR1-TCB is inactive. In contrast, killing of bronchial epithelial and renal cortical cells with low FOLR1 expression is prevented compared to the parental FOLR1-TCB. The findings are confirmed for mesothelin as alternative tumor antigen. Thus, masking the anti-CD3 Fab fragment with an anti-idiotypic mask and cleavage of the mask by tumor-specific proteases can be applied to enhance specificity and safety of TCBs.


Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , CD3 Complex/immunology , Folate Receptor 1/immunology , Peptide Hydrolases/metabolism , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/therapeutic use , Cell Line, Tumor , GPI-Linked Proteins/immunology , Humans , Immunotherapy , Lymphocyte Activation/drug effects , Mesothelin , Mice , Molecular Targeted Therapy , Xenograft Model Antitumor Assays
11.
Cancer Res ; 80(13): 2903-2913, 2020 07 01.
Article En | MEDLINE | ID: mdl-32409308

CD8-expressing T cells are the main effector cells in cancer immunotherapy. Treatment-induced changes in intratumoral CD8+ T cells may represent a biomarker to identify patients responding to cancer immunotherapy. Here, we have used a 89Zr-radiolabeled human CD8-specific minibody (89Zr-Df-IAB22M2C) to monitor CD8+ T-cell tumor infiltrates by PET. The ability of this tracer to quantify CD8+ T-cell tumor infiltrates was evaluated in preclinical studies following single-agent treatment with FOLR1-T-cell bispecific (TCB) antibody and combination therapy of CEA-TCB (RG7802) and CEA-targeted 4-1BB agonist CEA-4-1BBL. In vitro cytotoxicity assays with peripheral blood mononuclear cells and CEA-expressing MKN-45 gastric or FOLR1-expressing HeLa cervical cancer cells confirmed noninterference of the anti-CD8-PET-tracer with the mode of action of CEA-TCB/CEA-4-1BBL and FOLR1-TCB at relevant doses. In vivo, the extent of tumor regression induced by combination treatment with CEA-TCB/CEA-4-1BBL in MKN-45 tumor-bearing humanized mice correlated with intratumoral CD8+ T-cell infiltration. This was detectable by 89Zr-IAB22M2C-PET and γ-counting. Similarly, single-agent treatment with FOLR1-TCB induced strong CD8+ T-cell infiltration in HeLa tumors, where 89Zr-Df-IAB22M2C again was able to detect CD8 tumor infiltrates. CD8-IHC confirmed the PET imaging results. Taken together, the anti-CD8-minibody 89Zr-Df-IAB22M2C revealed a high sensitivity for the detection of intratumoral CD8+ T-cell infiltrates upon either single or combination treatment with TCB antibody-based fusion proteins. These results provide further evidence that the anti-CD8 tracer, which is currently in clinical phase II, is a promising monitoring tool for intratumoral CD8+ T cells in patients treated with cancer immunotherapy. SIGNIFICANCE: Monitoring the pharmacodynamic activity of cancer immunotherapy with novel molecular imaging tools such as 89Zr-Df-IAB22M2C for PET imaging is of prime importance to identify patients responding early to cancer immunotherapy.


Antibodies, Bispecific/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Molecular Imaging/methods , Positron-Emission Tomography/methods , Uterine Cervical Neoplasms/immunology , Zirconium/metabolism , Animals , Antibodies, Bispecific/immunology , Carcinoembryonic Antigen , Female , Folate Receptor 1/immunology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Radiopharmaceuticals/metabolism , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/therapy
12.
Gynecol Oncol ; 157(2): 379-385, 2020 05.
Article En | MEDLINE | ID: mdl-32081463

PURPOSE: To evaluate the safety and clinical activity of mirvetuximab soravtansine, an antibody-drug conjugate comprising a humanized anti-folate receptor alpha (FRα) monoclonal antibody, cleavable linker, and the maytansinoid DM4, a potent tubulin-targeting agent, in combination with bevacizumab in patients with FRα-positive, platinum-resistant ovarian cancer. METHODS: Patients with platinum-resistant epithelial ovarian, fallopian tube, or primary peritoneal cancer were administered mirvetuximab soravtansine (6 mg/kg, adjusted ideal body weight) and bevacizumab (15 mg/kg) once every 3 weeks. Eligibility included FRα positivity by immunochemistry and prior bevacizumab exposure was permitted. Adverse events, tumor response, and progression-free survival (PFS) were determined. RESULTS: Sixty-six patients, with a median of 3 prior lines of therapy (range, 1-8), received the combination of mirvetuximab soravtansine and bevacizumab at full dosing during the escalation and expansion stages of the study. Adverse events were generally mild-to-moderate (≤grade 2) with diarrhea, blurred vision, nausea, and fatigue being the most common treatment-related toxicities. Six cases of pneumonitis (9%; all grade 1 or 2), an adverse event of special interest, were observed. The confirmed objective response rate (ORR) was 39%, including 5 complete responses and 21 partial responses, and the median PFS was 6.9 months. The combination was particularly active in the subset of patients (n = 16) who were bevacizumab-naïve, less heavily pretreated (1-2 prior lines), and whose tumors exhibited medium/high FRα expression (ORR, 56% with a median duration of response of 12 months; PFS, 9.9 months). CONCLUSION: The combination of mirvetuximab soravtansine with bevacizumab is well tolerated in patients with platinum-resistant, recurrent ovarian cancer. The encouraging efficacy measures compare favorably to reported outcomes for bevacizumab combined with standard chemotherapy in similar patient populations.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/administration & dosage , Bevacizumab/adverse effects , Carcinoma, Ovarian Epithelial/immunology , Drug Resistance, Neoplasm , Female , Folate Receptor 1/immunology , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Maytansine/administration & dosage , Maytansine/adverse effects , Maytansine/analogs & derivatives , Middle Aged , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/immunology , Progression-Free Survival
13.
Front Immunol ; 10: 1994, 2019.
Article En | MEDLINE | ID: mdl-31497024

T-cell engaging bispecific antibodies (biAbs) can mediate potent and specific tumor cell eradication in liquid cancers. Substantial effort has been invested in expanding this concept to solid cancers. To explore their utility in the treatment of ovarian cancer, we built a set of asymmetric biAbs in IgG1-like format that bind CD3 on T cells with a conventional scFv arm and folate receptor 1 (FOLR1) on ovarian cancer cells with a conventional or a chemically programmed Fab arm. For avidity engineering, we also built an asymmetric biAb format with a tandem Fab arm. We show that both conventional and chemically programmed CD3 × FOLR1 biAbs exert specific in vitro and in vivo cytotoxicity toward FOLR1-expressing ovarian cancer cells by recruiting and activating T cells. While the conventional T-cell engaging biAb was curative in an aggressive mouse model of human ovarian cancer, the potency of the chemically programmed biAb was significantly boosted by avidity engineering. Both conventional and chemically programmed CD3 × FOLR1 biAbs warrant further investigation for ovarian cancer immunotherapy.


Antibodies, Bispecific/pharmacology , Folate Receptor 1/immunology , Ovarian Neoplasms/immunology , Animals , Antibodies, Bispecific/pharmacokinetics , Cell Line , Female , Humans , Lymphocyte Activation , Mice, Inbred BALB C , T-Lymphocytes/immunology
14.
Mol Pharm ; 16(9): 3996-4006, 2019 09 03.
Article En | MEDLINE | ID: mdl-31369274

Folate receptor α (FRα) is a well-studied tumor biomarker highly expressed in many epithelial tumors such as breast, ovarian, and lung cancers. Mirvetuximab soravtansine (IMGN853) is the antibody-drug conjugate of FRα-binding humanized monoclonal antibody M9346A and cytotoxic maytansinoid drug DM4. IMGN853 is currently being evaluated in multiple clinical trials, in which the immunohistochemical evaluation of an archival tumor or biopsy specimen is used for patient screening. However, limited tissue collection may lead to inaccurate diagnosis due to tumor heterogeneity. Herein, we developed a zirconium-89 (89Zr)-radiolabeled M9346A (89Zr-M9346A) as an immuno-positron emission tomography (immuno-PET) radiotracer to evaluate FRα expression in triple-negative breast cancer (TNBC) patients, providing a novel means to guide intervention with therapeutic IMGN853. In this study, we verified the binding specificity and immunoreactivity of 89Zr-M9346A by in vitro studies in FRαhigh cells (HeLa) and FRαlow cells (OVCAR-3). In vivo PET/computed tomography (PET/CT) imaging in HeLa xenografts and TNBC patient-derived xenograft (PDX) mouse models with various levels of FRα expression demonstrated its targeting specificity and sensitivity. Following PET imaging, the treatment efficiencies of IMGN853, pemetrexed, IMGN853 + pemetrexed, paclitaxel, and saline were assessed in FRαhigh and FRαlow TNBC PDX models. The correlation between 89Zr-M9346A tumor uptake and treatment response using IMGN853 in FRαhigh TNBC PDX model suggested the potential of 89Zr-M9346A PET as a noninvasive tool to prescreen patients based on the in vivo PET imaging for IMGN853-targeted treatment.


Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Folate Receptor 1/immunology , Folate Receptor 1/metabolism , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Maytansine/analogs & derivatives , Radioisotopes/pharmacokinetics , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/drug therapy , Zirconium/pharmacokinetics , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Drug Therapy, Combination , Female , HeLa Cells , Humans , Immunoconjugates/chemistry , Male , Maytansine/chemistry , Maytansine/pharmacokinetics , Maytansine/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Molecular Targeted Therapy/methods , Paclitaxel/therapeutic use , Pemetrexed/therapeutic use , Positron Emission Tomography Computed Tomography/methods , Radioisotopes/chemistry , Tissue Distribution , Treatment Outcome , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays , Zirconium/chemistry
15.
Cytokine Growth Factor Rev ; 45: 45-52, 2019 02.
Article En | MEDLINE | ID: mdl-30770191

The glycoprotein FRα is a membrane-attached transport protein that is shielded from the immune system in healthy cells. However, it is upregulated in various malignancies, involved in cancer development and is also immunogenic. Furthermore, FRα is a tumor-associated antigen endowed with unique properties, thus rendering it a suitable target for immunotherapeutic development in cancer. Various anti- FRα immunotherapeutic strategies are thus currently being developed and clinically assessed for the treatment of various solid tumors. These approaches include passive anti-FRα immunotherapies, such as monoclonal antibodies, or active immunotherapies, such as CART, folate haptens and vaccines. In this review, we will explore the advances in the field of FRα-based immune therapies and discuss both their successes and shortcomings in the clinical setting.


Folate Receptor 1/antagonists & inhibitors , Folate Receptors, GPI-Anchored/immunology , Immunotherapy/methods , Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Clinical Trials as Topic , Female , Folate Receptor 1/immunology , Folate Receptors, GPI-Anchored/genetics , Humans , Mice , Neoplasms/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy
16.
J Clin Psychopharmacol ; 39(1): 28-38, 2019.
Article En | MEDLINE | ID: mdl-30566416

PURPOSE/BACKGROUND: Interventions for attention-deficit/hyperactivity disorder (ADHD) may be inadequate for some patients. There is evidence that supplementation with L-methylfolate augments antidepressant agent effects and thus might also augment ADHD treatment effects by a common catecholaminergic mechanism. METHODS: Forty-four adults with Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition diagnosis of ADHD participated in a randomized, double-blind, placebo-controlled, 12-week trial of 15 mg of L-methylfolate in combination with osmotic-release oral system methylphenidate. Osmotic-release oral system methylphenidate was dose optimized over the first 6 weeks. We evaluated the effects on ADHD symptoms, self-report on the Behavior Rating Inventory of Executive Function of executive function, methylphenidate dosing, neuropsychological test measures, the Adult ADHD Self-report scale, emotional dysregulation, social adjustment, and work productivity, as well as moderating effects of body mass index, autoantibodies to folate receptors, and select genetic polymorphisms. RESULTS: L-Methylfolate was well tolerated, with no significant effect over placebo except improvement from abnormal measures on the mean adaptive dimension of the ASR scale (χ = 4.36, P = 0.04). Methylphenidate dosing was significantly higher in individuals on L-methylfolate over time (χ = 7.35, P = 0.007). Exploratory analyses suggested that variation in a guanosine triphosphate cyclohydrolase gene predicted association with higher doses of methylphenidate (P < 0.001). CONCLUSIONS: L-Methylfolate was associated with no change in efficacy on measures relevant to neuropsychiatric function in adults with ADHD, other than suggestion of reduced efficacy of methylphenidate. Further investigation would be required to confirm this effect and its mechanism and the genotype prediction of effects on dosing.


Attention Deficit Disorder with Hyperactivity/drug therapy , Methylphenidate/therapeutic use , Tetrahydrofolates/therapeutic use , Administration, Oral , Adult , Attention Deficit Disorder with Hyperactivity/genetics , Autoantibodies/blood , Autoantibodies/immunology , Central Nervous System Stimulants/therapeutic use , Delayed-Action Preparations/therapeutic use , Diet Therapy , Dietary Supplements , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination , Executive Function/drug effects , Female , Folate Receptor 1/immunology , GTP Cyclohydrolase/genetics , Humans , Male , Methylphenidate/administration & dosage , Neuropsychological Tests , Pilot Projects , Tetrahydrofolates/adverse effects , Treatment Outcome , Young Adult
17.
Biosci Trends ; 12(3): 298-308, 2018 Jul 17.
Article En | MEDLINE | ID: mdl-29899195

Our goal is to develop a switch-controlled approach to enable better control of reactivity and safety of chimeric antigen receptor (CAR)-T therapy for non-small-cell lung cancer (NSCLC). Lentiviral transduction was performed to generate anti-FITC CAR-T cells and target cells stably expressing either isoform of the folate receptor. Colorimetric-based cytotoxic assay, enzyme-linked immunosorbent assay, and multiparametric flow cytometry analysis were used to evaluate the specificity and activity of CAR-T cells in vitro. Human primary T cells stably expressing the fully human anti-FITC CAR were generated. Anti-FITC CAR-T cells displayed antigen-specific and folate-FTIC dependent reactivity against engineered A549-FRα and THP-1-FRß. The selective activation and proliferation of anti-FITC CAR-T cells in vitro stringently relied on the co-existence of folate-FITC and FR- expressing target cells and was dose-titratable with the folate-FITC switch. The excellent in vitro efficacy and specificity of an adaptor-controlled CAR-T therapy to target both tumor cells and tumor-associated macrophages in NSCLCs were validated.


Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , A549 Cells , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Flow Cytometry , Folate Receptor 1/immunology , Folate Receptor 1/metabolism , HEK293 Cells , HL-60 Cells , Humans , Jurkat Cells , Lentivirus/genetics , Ligands , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophages/immunology , T-Lymphocytes/immunology , Transduction, Genetic , Tumor Microenvironment/immunology
18.
PLoS One ; 13(6): e0198347, 2018.
Article En | MEDLINE | ID: mdl-29874279

Gastric cancer is a malignancy that has a high mortality rate. Although progress has been made in the treatment of gastric cancer, many patients experience cancer recurrence and metastasis. Folate receptor 1 (FOLR1) is overexpressed on the cell surface in over one-third of gastric cancer patients, but rarely is expressed in normal tissue. This makes FOLR1 a potential target for chimeric antigen receptor (CAR) T cell immunotherapy, although the function of FOLR1 has not been elucidated. CAR are engineered fusion receptor composed of an antigen recognition region and signaling domains. T cells expressing CAR have specific activation and cytotoxic effects against cancer cells containing the target antigen. In this study, we generated a CAR that targets FOLR1 composed of a single-chain variable fragment (scFv) of FOLR1 antibody and signaling domains consisting of CD28 and CD3ζ. Both FOLR1-CAR KHYG-1, a natural killer cell line, and FOLR1-CAR T cells recognized FOLR1-positive gastric cancer cells in a MHC-independent manner and induced secretion of various cytokines and caused cell death. Conclusively, this is the first study to demonstrate that CAR KHYG-1/T cells targeting FOLR1 are effective against FOLR1-positive gastric cancer cells.


Folate Receptor 1/immunology , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Stomach Neoplasms/therapy , T-Lymphocytes/transplantation , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Jurkat Cells , K562 Cells , Mice , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Stomach Neoplasms/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
19.
Allergy ; 73(12): 2328-2341, 2018 12.
Article En | MEDLINE | ID: mdl-29654623

BACKGROUND: Designing biologically informative models for assessing the safety of novel agents, especially for cancer immunotherapy, carries substantial challenges. The choice of an in vivo system for studies on IgE antibodies represents a major impediment to their clinical translation, especially with respect to class-specific immunological functions and safety. Fcε receptor expression and structure are different in humans and mice, so that the murine system is not informative when studying human IgE biology. By contrast, FcεRI expression and cellular distribution in rats mirror that of humans. METHODS: We are developing MOv18 IgE, a human chimeric antibody recognizing the tumour-associated antigen folate receptor alpha. We created an immunologically congruent surrogate rat model likely to recapitulate human IgE-FcεR interactions and engineered a surrogate rat IgE equivalent to MOv18. Employing this model, we examined in vivo safety and efficacy of antitumour IgE antibodies. RESULTS: In immunocompetent rats, rodent IgE restricted growth of syngeneic tumours in the absence of clinical, histopathological or metabolic signs associated with obvious toxicity. No physiological or immunological evidence of a "cytokine storm" or allergic response was seen, even at 50 mg/kg weekly doses. IgE treatment was associated with elevated serum concentrations of TNFα, a mediator previously linked with IgE-mediated antitumour and antiparasitic functions, alongside evidence of substantially elevated tumoural immune cell infiltration and immunological pathway activation in tumour-bearing lungs. CONCLUSION: Our findings indicate safety of MOv18 IgE, in conjunction with efficacy and immune activation, supporting the translation of this therapeutic approach to the clinical arena.


Antibodies, Monoclonal, Murine-Derived/adverse effects , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Immunoglobulin E/adverse effects , Immunoglobulin E/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Receptors, IgE/metabolism , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/metabolism , Cell Line, Tumor , Folate Receptor 1/immunology , Humans , Immunoglobulin E/administration & dosage , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Mice , Models, Animal , Neoplasms/pathology , Protein Binding , Rats , Statistics, Nonparametric , Treatment Outcome , Tumor Necrosis Factor-alpha/blood
20.
Mol Genet Metab ; 124(1): 87-93, 2018 05.
Article En | MEDLINE | ID: mdl-29661558

INTRODUCTION: Cerebral folate deficiency (CFD) syndromes are defined as neuro-psychiatric conditions with low CSF folate and attributed to different causes such as autoantibodies against the folate receptor-alpha (FR) protein that can block folate transport across the choroid plexus, FOLR1 gene mutations or mitochondrial disorders. High-dose folinic acid treatment restores many neurologic deficits. STUDY AIMS AND METHODS: Among 36 patients from 33 families the infantile-onset CFD syndrome was diagnosed based on typical clinical features and low CSF folate. All parents were healthy. Three families had 2 affected siblings, while parents from 4 families were first cousins. We analysed serum FR autoantibodies and the FOLR1 and FOLR2 genes. Among three consanguineous families homozygosity mapping attempted to identify a monogenetic cause. Whole exome sequencing (WES) was performed in the fourth consanguineous family, where two siblings also suffered from polyneuropathy as an atypical finding. RESULTS: Boys (72%) outnumbered girls (28%). Most patients (89%) had serum FR autoantibodies fluctuating over 5-6 weeks. Two children had a genetic FOLR1 variant without pathological significance. Homozygosity mapping failed to detect a single autosomal recessive gene. WES revealed an autosomal recessive polynucleotide kinase 3´phosphatase (PNKP) gene abnormality in the siblings with polyneuropathy. DISCUSSION: Infantile-onset CFD was characterized by serum FR autoantibodies as its predominant pathology whereas pathogenic FOLR1 gene mutations were absent. Homozygosity mapping excluded autosomal recessive inheritance of any single responsible gene. WES in one consanguineous family identified a PNKP gene abnormality that explained the polyneuropathy and also its contribution to the infantile CFD syndrome because the PNKP gene plays a dual role in both neurodevelopment and immune-regulatory function. Further research for candidate genes predisposing to FRα-autoimmunity is suggested to include X-chromosomal and non-coding DNA regions.


Autoantibodies/blood , Brain Diseases, Metabolic, Inborn/genetics , Folate Receptor 1/immunology , Folic Acid Deficiency/genetics , Adolescent , Brain Diseases, Metabolic, Inborn/cerebrospinal fluid , Brain Diseases, Metabolic, Inborn/diagnosis , Child , Child, Preschool , Consanguinity , DNA Repair Enzymes/genetics , Diagnosis, Differential , Family , Female , Folate Receptor 1/genetics , Folate Receptor 2/genetics , Folic Acid/cerebrospinal fluid , Folic Acid Deficiency/cerebrospinal fluid , Folic Acid Deficiency/diagnosis , Humans , Infant , Male , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polyneuropathies/etiology , Exome Sequencing , Young Adult
...