Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 23(2): 187-198, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37828725

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a solid tumor type that arises in the squamous epithelial cells lining the mucosal surfaces of the upper aerodigestive tract. Long-term survival of patients with advanced disease stage remains disappointing with current treatment options. We show that tissue factor is abundantly expressed on patient-derived HNSCC cell lines, xenograft tumor material, and tumor biopsies from patients with HNSCC. Tisotumab vedotin (TV) is an antibody-drug conjugate (ADC) directed to tissue factor, a protein expressed in many solid tumors. HNSCC cells and xenograft tumors were efficiently eliminated in vitro and in vivo with TV-monotherapy compared with treatment with a control antibody conjugated to monomethyl auristatin E (MMAE). Antitumor activity of TV was also tested in vivo in combination with chemoradiotherapy, standard of care for patients with advanced stage HNSCC tumors outside the oral cavity. Preclinical studies showed that by adding TV to chemoradiotherapy, survival was markedly improved, and TV, not radiotherapy or chemotherapy, was the main driver of antitumor activity. Interestingly, TV-induced cell death in xenograft tumors showed an influx of macrophages indicative of a potential immune-mediated mode-of-action. In conclusion, on the basis of these preclinical data, TV may be a novel treatment modality for patients suffering from head and neck cancer and is hypothesized to improve efficacy of chemoradiotherapy. SIGNIFICANCE: This work shows preclinical in vitro and in vivo antitumor activity of the antibody-drug conjugate Tisotumab vedotin in head and neck cancer models, and enhanced activity in combination with chemoradiotherapy, supporting further clinical development for this cancer type.


Subject(s)
Head and Neck Neoplasms , Immunoconjugates , Humans , Cell Line, Tumor , Chemoradiotherapy , Head and Neck Neoplasms/drug therapy , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy , Thromboplastin , Xenograft Model Antitumor Assays , Animals
2.
Cancer Rep (Hoboken) ; 6(2): e1699, 2023 02.
Article in English | MEDLINE | ID: mdl-36806722

ABSTRACT

BACKGROUND: Elevated tissue factor (TF) expression, although restricted in normal tissue, has been reported in multiple solid cancers, and expression has been associated with poor prognosis. This manuscript compares TF expression across various solid tumor types via immunohistochemistry in a single study, which has not been performed previously. AIMS: To increase insight in the prevalence and cellular localization of TF expression across solid cancer types, we performed a detailed and systematic analysis of TF expression in tumor tissue obtained from patients with ovarian, esophageal, bladder, cervical, endometrial, pancreatic, prostate, colon, breast, non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC), and glioblastoma. The spatial and temporal variation of TF expression was analyzed over time and upon disease progression in patient-matched biopsies taken at different timepoints. In addition, TF expression in patient-matched primary tumor and metastatic lesions was also analyzed. METHODS AND RESULTS: TF expression was detected via immunohistochemistry (IHC) using a validated TF-specific antibody. TF was expressed in all cancer types tested, with highest prevalence in pancreatic cancer, cervical cancer, colon cancer, glioblastoma, HNSCC, and NSCLC, and lowest in breast cancer. Staining was predominantly membranous in pancreatic, cervical, and HNSCC, and cytoplasmic in glioblastoma and bladder cancer. In general, expression was consistent between biopsies obtained from the same patient over time, although variability was observed for individual patients. NSCLC biopsies of primary tumor and matched lymph node metastases showed no clear difference in TF expression overall, although individual patient changes were observed. CONCLUSION: This study shows that TF is expressed across a broad range of solid cancer types, and expression is present upon tumor dissemination and over the course of treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Glioblastoma , Head and Neck Neoplasms , Lung Neoplasms , Male , Female , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Squamous Cell Carcinoma of Head and Neck , Thromboplastin/analysis , Lung Neoplasms/pathology
3.
Life Sci Alliance ; 5(11)2022 Nov.
Article in English | MEDLINE | ID: mdl-36271507

ABSTRACT

CD3 bispecific antibodies (bsAbs) show great promise as anticancer therapeutics. Here, we show in-depth mechanistic studies of a CD3 bsAb in solid cancer, using DuoBody-CD3x5T4. Cross-linking T cells with tumor cells expressing the oncofetal antigen 5T4 was required to induce cytotoxicity. Naive and memory CD4+ and CD8+ T cells were equally effective at mediating cytotoxicity, and DuoBody-CD3x5T4 induced partial differentiation of naive T-cell subsets into memory-like cells. Tumor cell kill was associated with T-cell activation, proliferation, and production of cytokines, granzyme B, and perforin. Genetic knockout of FAS or IFNGR1 in 5T4+ tumor cells abrogated tumor cell kill. In the presence of 5T4+ tumor cells, bystander kill of 5T4- but not of 5T4-IFNGR1- tumor cells was observed. In humanized xenograft models, DuoBody-CD3x5T4 antitumor activity was associated with intratumoral and peripheral blood T-cell activation. Lastly, in dissociated patient-derived tumor samples, DuoBody-CD3x5T4 activated tumor-infiltrating lymphocytes and induced tumor-cell cytotoxicity, even when most tumor-infiltrating lymphocytes expressed PD-1. These data provide an in-depth view on the mechanism of action of a CD3 bsAb in preclinical models of solid cancer.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Antibodies, Bispecific/pharmacology , CD8-Positive T-Lymphocytes , Granzymes/pharmacology , CD3 Complex/pharmacology , Cytotoxicity, Immunologic , Perforin/pharmacology , Programmed Cell Death 1 Receptor , Neoplasms/drug therapy , Cytokines
4.
Cancer Res ; 81(7): 1775-1787, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33531370

ABSTRACT

Although immune checkpoint blockade (ICB) has shown remarkable clinical benefit in a subset of patients with melanoma and lung cancer, most patients experience no durable benefit. The receptor tyrosine kinase AXL is commonly implicated in therapy resistance and may serve as a marker for therapy-refractory tumors, for example in melanoma, as we previously demonstrated. Here, we show that enapotamab vedotin (EnaV), an antibody-drug conjugate targeting AXL, effectively targets tumors that display insensitivity to immunotherapy or tumor-specific T cells in several melanoma and lung cancer models. In addition to its direct tumor cell killing activity, EnaV treatment induced an inflammatory response and immunogenic cell death in tumor cells and promoted the induction of a memory-like phenotype in cytotoxic T cells. Combining EnaV with tumor-specific T cells proved superior to either treatment alone in models of melanoma and lung cancer and induced ICB benefit in models otherwise insensitive to anti-PD-1 treatment. Our findings indicate that targeting AXL-expressing, immunotherapy-resistant tumors with EnaV causes an immune-stimulating tumor microenvironment and enhances sensitivity to ICB, warranting further investigation of this treatment combination. SIGNIFICANCE: These findings show that targeting AXL-positive tumor fractions with an antibody-drug conjugate enhances antitumor immunity in several humanized tumor models of melanoma and lung cancer.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunoconjugates/therapeutic use , Lung Neoplasms/therapy , Melanoma/therapy , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance, Neoplasm/immunology , Drug Synergism , HEK293 Cells , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immunoconjugates/administration & dosage , Immunotherapy , Lung Neoplasms/pathology , Male , Melanoma/pathology , Mice , Mice, Nude , Mice, Transgenic , Molecular Targeted Therapy/methods , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
5.
Mol Cancer Ther ; 18(2): 312-322, 2019 02.
Article in English | MEDLINE | ID: mdl-30381448

ABSTRACT

Immunotherapy of cancer with CD3-targeting bispecific antibodies (CD3 bsAb) is a fast developing field, and multiple tumor-associated antigens (TAA) are evaluated for hematologic and solid malignancies. The efficacy of these CD3 bsAb is usually examined in xenograft mouse tumor models with human T cells or in genetically engineered mouse models, where human TAA are introduced. These models often fail to fully recapitulate the natural tumor environment, especially for solid cancers, because of interspecies differences. Here, we investigated the systemic and intratumoral effects of a mouse CD3 bsAb in a fully immune-competent mouse melanoma model. Systemic administration of 0.5 mg/kg antibody induced a brief overall T-cell activation that was selectively sustained in the tumor microenvironment for several days. A fast subsequent influx of inflammatory macrophages into the tumor microenvironment was observed, followed by an increase in the number of CD4+ and CD8+ T cells. Although the capacity to directly kill melanoma cells in vitro was very modest, optimal tumor elimination was observed in vivo, even in the absence of CD8+ T cells, implying a redundancy in T-cell subsets for therapeutic efficacy. Finally, we took advantage of the full immune competence of our mouse model and tested immune memory induction. Despite a strong initial immunity against melanoma, treatment with the CD3 bsAb did not install protective memory responses. The observed mechanisms of action revealed in this immune-competent mouse model might form a rational basis for combinatorial approaches.


Subject(s)
Antibodies, Bispecific/administration & dosage , CD3 Complex/antagonists & inhibitors , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Melanoma/drug therapy , Animals , Antibodies, Bispecific/pharmacology , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Female , Humans , Immunologic Memory , Lymphocyte Activation , Melanoma/immunology , Mice , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
6.
J Nucl Med ; 55(5): 830-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24686779

ABSTRACT

UNLABELLED: Longitudinal imaging of intratumoral distributions of antibodies in vivo in mouse cancer models is of great importance for developing cancer therapies. In this study, multipinhole SPECT with sub-half-millimeter resolution was tested for exploring intratumoral distributions of radiolabeled antibodies directed toward the epidermal growth factor receptor (EGFr) and compared with full 3-dimensional target expression assessed by immunohistochemistry. METHODS: (111)In-labeled zalutumumab, a human monoclonal human EGFr-targeting antibody, was administered at a nonsaturating dose to 3 mice with xenografted A431 tumors exhibiting high EGFr expression. Total-body and focused in vivo tumor SPECT was performed at 0 and 48 h after injection and compared both visually and quantitatively with full 3-dimensional immunohistochemical staining for EGFr target expression. RESULTS: SPECT at 48 h after injection showed that activity was predominantly concentrated in the tumor (10.5% ± 1.3% of the total-body activity; average concentration, 30.1% ± 4.6% of the injected dose per cubic centimeter). (111)In-labeled EGFr-targeting antibodies were distributed heterogeneously throughout the tumor. Some hot spots were observed near the tumor rim. Immunohistochemistry indicated that the antibody distributions obtained by SPECT were morphologically similar to those obtained for ex vivo EGFr target expression. Regions showing low SPECT activity were necrotic or virtually negative for EGFr target expression. A good correlation (r = 0.86, P < 0.0001) was found between the percentage of regions showing low activity on SPECT and the percentage of necrotic tissue on immunohistochemistry. CONCLUSION: Multipinhole SPECT enables high-resolution visualization and quantification of the heterogeneity of (111)In-zalutumumab concentrations in vivo.


Subject(s)
Antibodies/chemistry , ErbB Receptors/metabolism , Imaging, Three-Dimensional , Tomography, Emission-Computed, Single-Photon , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Female , Humans , Image Processing, Computer-Assisted , Immunoconjugates/chemistry , Immunohistochemistry , Indium Radioisotopes/chemistry , Mice , Mice, SCID , Neoplasm Transplantation , Reproducibility of Results , Tomography, X-Ray Computed
7.
Cancer Res ; 74(4): 1214-26, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24371232

ABSTRACT

Tissue factor (TF) is aberrantly expressed in solid cancers and is thought to contribute to disease progression through its procoagulant activity and its capacity to induce intracellular signaling in complex with factor VIIa (FVIIa). To explore the possibility of using tissue factor as a target for an antibody-drug conjugate (ADC), a panel of human tissue factor-specific antibodies (TF HuMab) was generated. Three tissue factor HuMab, that induced efficient inhibition of TF:FVIIa-dependent intracellular signaling, antibody-dependent cell-mediated cytotoxicity, and rapid target internalization, but had minimal impact on tissue factor procoagulant activity in vitro, were conjugated with the cytotoxic agents monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF). Tissue factor-specific ADCs showed potent cytotoxicity in vitro and in vivo, which was dependent on tissue factor expression. TF-011-MMAE (HuMax-TF-ADC) was the most potent ADC, and the dominant mechanism of action in vivo was auristatin-mediated tumor cell killing. Importantly, TF-011-MMAE showed excellent antitumor activity in patient-derived xenograft (PDX) models with variable levels of tissue factor expression, derived from seven different solid cancers. Complete tumor regression was observed in all PDX models, including models that showed tissue factor expression in only 25% to 50% of the tumor cells. In conclusion, TF-011-MMAE is a promising novel antitumor agent with potent activity in xenograft models that represent the heterogeneity of human tumors, including heterogeneous target expression.


Subject(s)
Aminobenzoates/therapeutic use , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Oligopeptides/therapeutic use , Thromboplastin/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Cells, Cultured , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Thromboplastin/antagonists & inhibitors , Xenograft Model Antitumor Assays
8.
PLoS One ; 7(6): e39706, 2012.
Article in English | MEDLINE | ID: mdl-22761877

ABSTRACT

Epidermal growth factor receptor (EGFR) inhibitors are widely used in the treatment of cancer. EGFR-targeted treatment is known to be associated with a high incidence of dermatological adverse reactions, including papulopustular rash, which can be dose-limiting and may affect compliance to treatment. Currently, the pathways involved in EGFR inhibitor-induced rash are poorly understood and few treatment options for this adverse event are available. Here, we developed a model for induction of papulopustular rash in healthy human volunteers by subcutaneous injection of the anti-EGFR monoclonal antibody zalutumumab. The injection sites and surrounding skin were evaluated by a dermatologist for the presence or absence of papulopustular rash and skin biopsies were taken to confirm the macroscopical findings by immunohistochemistry. Locally injected zalutumumab induced a papulopustular rash, characterized by acute follicular neutrophil-rich hair follicle inflammation, and thus mimicked adverse events induced by systemic administration of EGFR inhibitors. In this model, we tested the hypothesis that neutrophils, attracted by IL-8, play a central role in the observed rash. Indeed, concomitant local repeat dose treatment with HuMab-10F8, a neutralizing human antibody against IL-8, reduced the rash. Inhibition of IL-8 can therefore ameliorate dermatological adverse events induced by treatment with EGFR inhibitors.


Subject(s)
Antibodies, Monoclonal/adverse effects , ErbB Receptors/antagonists & inhibitors , Interleukin-8/immunology , Neutralization Tests , Skin Diseases/prevention & control , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Humans , Injections, Subcutaneous , Skin Diseases/chemically induced , Skin Diseases/pathology
9.
Cancer Res ; 66(15): 7630-8, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885363

ABSTRACT

The epidermal growth factor receptor (EGFR) is overexpressed on many solid tumors and represents an attractive target for antibody therapy. Here, we describe the effect of receptor-mediated antibody internalization on the pharmacokinetics and dose-effect relationship of a therapeutic monoclonal antibody (mAb) against EGFR (2F8). This mAb was previously found therapeutically active in mouse tumor models by two dose-dependent mechanisms of action: blockade of ligand binding and induction of antibody-dependent cell-mediated cytotoxicity. In vitro studies showed 2F8 to be rapidly internalized by EGFR-overexpressing cells. In vivo, accelerated 2F8 clearance was observed in cynomolgus monkeys at low doses but not at high doses. This enhanced clearance seemed to be receptor dependent and was included in a pharmacokinetic model designed to explain its nonlinearity. Receptor-mediated clearance was also found to affect in situ antibody concentrations in tumor tissue. Ex vivo analyses of xenograft tumors of 2F8-treated nude mice revealed that relatively high antibody plasma concentrations were required for maximum EGFR saturation in high-EGFR-expressing human A431 tumors, in contrast to lower-EGFR-expressing human xenograft tumors. In summary, receptor-mediated antibody internalization and degradation provides a saturable route of clearance that significantly affects pharmacokinetics, particularly at low antibody doses. EGFR saturation in normal tissues does not predict saturation in tumor tissue as local antibody concentrations in EGFR-overexpressing tumors may be more rapidly reduced by antibody internalization. Consequently, antibody saturation of the receptor may be affected, thereby affecting the local mechanism of action.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , ErbB Receptors/immunology , Animals , Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/therapy , Dose-Response Relationship, Immunologic , ErbB Receptors/biosynthesis , Female , Humans , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Xenograft Model Antitumor Assays
10.
J Immunol ; 173(7): 4699-707, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383606

ABSTRACT

Epidermal growth factor receptor (EGF-R) overexpression is common in a large number of solid tumors and represents a negative prognostic indicator. Overexpression of EGF-R is strongly tumor associated, and this tyrosine kinase type receptor is considered an attractive target for Ab therapy. In this study, we describe the evaluation of mAb 2F8, a high avidity human mAb (IgG1kappa) directed against EGF-R, developed using human Ig transgenic mice. mAb 2F8 effectively blocked binding of EGF and TGF-alpha to the EGF-R. At saturating concentrations, 2F8 completely blocked EGF-R signaling and inhibited the in vitro proliferation of EGF-R-overexpressing A431 cells. At much lower concentrations, associated with low receptor occupancy, 2F8 induced efficient Ab-dependent cell-mediated cytotoxicity (ADCC) in vitro. In vivo studies showed potent antitumor effects in models with A431 tumor xenografts in athymic mice. Ex vivo analysis of the EGF-R status in tumor xenografts in 2F8-treated mice revealed that there are two therapeutic mechanisms. First, blocking of EGF-R signaling, which is most effective at complete receptor saturation and therefore requires a relatively high Ab dose. Second, at very low 2F8 receptor occupancy, we observed potent antitumor effects in mice, which are likely based on the engagement of immune effector mechanisms, in particular ADCC. Taken together, our findings indicate that ADCC represents an important effector mechanism of this Ab, which is effective at relatively low dose.


Subject(s)
Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Binding Sites, Antibody , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Immunologic , ErbB Receptors/physiology , Female , Growth Inhibitors/therapeutic use , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/pathology , Neoplasms/prevention & control , Protein Binding , Signal Transduction/immunology , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...