Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Ther Oncol ; 32(1): 200758, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38596304

ABSTRACT

Oncolytic viruses are engineered to selectively kill tumor cells and have demonstrated promising results in early-phase clinical trials. To further modulate the innate and adaptive immune system, we generated AZD4820, a vaccinia virus engineered to express interleukin-12 (IL-12), a potent cytokine involved in the activation of natural killer (NK) and T cells and the reprogramming of the tumor immune microenvironment. Testing in cultured human tumor cell lines demonstrated broad in vitro oncolytic activity and IL-12 transgene expression. A surrogate virus expressing murine IL-12 demonstrated antitumor activity in both MC38 and CT26 mouse syngeneic tumor models that responded poorly to immune checkpoint inhibition. In both models, AZD4820 significantly upregulated interferon-gamma (IFN-γ) relative to control mice treated with oncolytic vaccinia virus (VACV)-luciferase. In the CT26 study, 6 of 10 mice had a complete response after treatment with AZD4820 murine surrogate, whereas control VACV-luciferase-treated mice had 0 of 10 complete responders. AZD4820 treatment combined with anti-PD-L1 blocking antibody augmented tumor-specific T cell immunity relative to monotherapies. These findings suggest that vaccinia virus delivery of IL-12, combined with immune checkpoint blockade, elicits antitumor immunity in tumors that respond poorly to immune checkpoint inhibitors.

2.
Clin Cancer Res ; 29(6): 1086-1101, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36355054

ABSTRACT

PURPOSE: We evaluated the activity of AZD8205, a B7-H4-directed antibody-drug conjugate (ADC) bearing a novel topoisomerase I inhibitor (TOP1i) payload, alone and in combination with the PARP1-selective inhibitor AZD5305, in preclinical models. EXPERIMENTAL DESIGN: IHC and deep-learning-based image analysis algorithms were used to assess prevalence and intratumoral heterogeneity of B7-H4 expression in human tumors. Several TOP1i-ADCs, prepared with Val-Ala or Gly-Gly-Phe-Gly peptide linkers, with or without a PEG8 spacer, were compared in biophysical, in vivo efficacy, and rat toxicology studies. AZD8205 mechanism of action and efficacy studies were conducted in human cancer cell line and patient-derived xenograft (PDX) models. RESULTS: Evaluation of IHC-staining density on a per-cell basis revealed a range of heterogeneous B7-H4 expression across patient tumors. This informed selection of bystander-capable Val-Ala-PEG8-TOP1i payload AZ14170133 and development of AZD8205, which demonstrated improved stability, efficacy, and safety compared with other linker-payload ADCs. In a study of 26 PDX tumors, single administration of 3.5 mg/kg AZD8205 provided a 69% overall response rate, according to modified RECIST criteria, which correlated with homologous recombination repair (HRR) deficiency (HRD) and elevated levels of B7-H4 in HRR-proficient models. Addition of AZD5305 sensitized very low B7-H4-expressing tumors to AZD8205 treatment, independent of HRD status and in models representing clinically relevant mechanisms of PARPi resistance. CONCLUSIONS: These data provide evidence for the potential utility of AZD8205 for treatment of B7-H4-expressing tumors and support the rationale for an ongoing phase 1 clinical study (NCT05123482). See related commentary by Pommier and Thomas, p. 991.


Subject(s)
Immunoconjugates , Neoplasms , Rats , Humans , Animals , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Topoisomerase I Inhibitors , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly (ADP-Ribose) Polymerase-1/genetics
3.
Cytotherapy ; 24(7): 720-732, 2022 07.
Article in English | MEDLINE | ID: mdl-35570170

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR) T cell therapy has yielded impressive clinical results in hematological malignancies and is a promising approach for solid tumor treatment. However, toxicity, including cytokine-release syndrome (CRS) and neurotoxicity, is a concern hampering its broader use. METHODS: In selecting a lead CAR-T candidate against the oncofetal antigen glypican 3 (GPC3), we compared CARs bearing a low- and high-affinity single-chain variable fragment (scFv) binding to a similar epitope and cross-reactive with murine GPC3. RESULTS: Where the high-affinity CAR-T cells were toxic in vivo, the low-affinity CAR maintained cytotoxic function against antigen-positive tumor cells but did not show toxicity against normal tissues. High-affinity CAR-induced toxicity was caused by on-target, off-tumor binding, based on the observation that higher doses of the high-affinity CAR-T caused toxicity in non-tumor-bearing mice and accumulated in organs with low expression of GPC3. To explore another layer of controlling CAR-T toxicity, we developed a means to target and eliminate CAR-T cells using anti-TNF-α antibody therapy after CAR-T infusion. The antibody was shown to function by eliminating early antigen-activated, but not all, CAR-T cells, allowing a margin where the toxic response could be effectively decoupled from antitumor efficacy with only a minor loss in tumor control. By exploring additional traits of the CAR-T cells after activation, we identified a mechanism whereby we could use approved therapeutics and apply them as an exogenous kill switch that eliminated early activated CAR-T following antigen engagement in vivo. CONCLUSIONS: By combining the reduced-affinity CAR with this exogenous control mechanism, we provide evidence that we can modulate and control CAR-mediated toxicity.


Subject(s)
Glypicans , Receptors, Chimeric Antigen , Animals , Cell Line, Tumor , Glypicans/metabolism , Immunotherapy, Adoptive/methods , Mice , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes , Tumor Necrosis Factor Inhibitors , Xenograft Model Antitumor Assays
4.
Nat Commun ; 10(1): 5167, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727888

ABSTRACT

BRAF and MEK1/2 inhibitors are effective in melanoma but resistance inevitably develops. Despite increasing the abundance of pro-apoptotic BIM and BMF, ERK1/2 pathway inhibition is predominantly cytostatic, reflecting residual pro-survival BCL2 family activity. Here, we show that uniquely low BCL-XL expression in melanoma biases the pro-survival pool towards MCL1. Consequently, BRAF or MEK1/2 inhibitors are synthetic lethal with the MCL1 inhibitor AZD5991, driving profound tumour cell death that requires BAK/BAX, BIM and BMF, and inhibiting tumour growth in vivo. Combination of ERK1/2 pathway inhibitors with BCL2/BCL-w/BCL-XL inhibitors is stronger in CRC, correlating with a low MCL1:BCL-XL ratio; indeed the MCL1:BCL-XL ratio is predictive of ERK1/2 pathway inhibitor synergy with MCL1 or BCL2/BCL-w/BCL-XL inhibitors. Finally, AZD5991 delays acquired BRAFi/MEKi resistance and enhances the efficacy of an ERK1/2 inhibitor in a model of acquired BRAFi + MEKi resistance. Thus combining ERK1/2 pathway inhibitors with MCL1 antagonists in melanoma could improve therapeutic index and patient outcomes.


Subject(s)
Apoptosis , MAP Kinase Signaling System , Melanoma/pathology , Molecular Targeted Therapy , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , MAP Kinase Signaling System/drug effects , Macrocyclic Compounds/pharmacology , Mice , Proto-Oncogene Proteins B-raf/metabolism , bcl-X Protein/metabolism
5.
Mol Cancer Ther ; 17(10): 2176-2186, 2018 10.
Article in English | MEDLINE | ID: mdl-30065100

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a membrane-bound glutamate carboxypeptidase that is highly expressed in nearly all prostate cancers with the highest expression in metastatic castration-resistant prostate cancer (mCRPC). The prevalence of increased surface expression and constitutive internalization of PSMA make it an attractive target for an antibody-drug conjugate (ADC) approach to treating patients with mCRPC. MEDI3726 (previously known as ADCT-401) is an ADC consisting of an engineered version of the anti-PSMA antibody J591 site specifically conjugated to the pyrrolobenzodiazepine (PBD) dimer tesirine. MEDI3726 specifically binds the extracellular domain of PSMA and, once internalized, releases the PBD dimer to crosslink DNA and trigger cell death. In vitro, MEDI3726 demonstrated potent and specific cytotoxicity in a panel of PSMA-positive prostate cancer cell lines, consistent with internalization and DNA interstrand crosslinking. In vivo, MEDI3726 showed robust antitumor activity against the LNCaP and the castration-resistant CWR22Rv1 prostate cancer cell line xenografts. MEDI3726 also demonstrated durable antitumor activity in the PSMA-positive human prostate cancer patient-derived xenograft (PDX) LuCaP models. This activity correlated with increased phosphorylated Histone H2AX in tumor xenografts treated with MEDI3726. MEDI3726 is being evaluated in a phase I clinical trial as a treatment for patients with metastatic castrate-resistant prostate cancer (NCT02991911). Mol Cancer Ther; 17(10); 2176-86. ©2018 AACR.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Glutamate Carboxypeptidase II/antagonists & inhibitors , Immunoconjugates/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cell Line, Tumor , Cross Reactions/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression , Glutamate Carboxypeptidase II/genetics , Glutamate Carboxypeptidase II/metabolism , Humans , Immunohistochemistry , Macaca fascicularis , Male , Mice , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Mol Ther ; 25(8): 1917-1932, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28578991

ABSTRACT

Vesicular stomatitis virus encoding the IFNß transgene (VSV-IFNß) is a mediator of potent oncolytic activity and is undergoing clinical evaluation for the treatment of solid tumors. Emerging preclinical and clinical data suggest treatment of tumors with oncolytic viruses may sensitize tumors to checkpoint inhibitors and increase the anti-tumor immune response. New generations of immuno-oncology molecules including T cell agonists are entering clinical development and could be hypothesized to enhance the activity of oncolytic viruses, including VSV-IFNß. Here, we show that VSV-IFNß exhibits multiple mechanisms of action, including direct cell killing, stimulation of an innate immune response, recruitment of CD8 T cells, and depletion of T regulatory cells. Moreover, VSV-IFNß promotes the establishment of a CD8 T cell response to endogenous tumor antigens. Our data demonstrate a significant enhancement of anti-tumor function for VSV-IFNß when combined with checkpoint inhibitors, but not OX40 agonists. While the addition of checkpoint inhibitors to VSV-IFNß generated robust tumor growth inhibition, it resulted in no increase in viral replication, transgene expression, or immunophenotypic changes beyond treatment with VSV-IFNß alone. We hypothesize that tumor-specific T cells generated by VSV-IFNß retain activity due to a lack of immune exhaustion when checkpoint inhibitors were used.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Immunotherapy , Neoplasms/genetics , Neoplasms/immunology , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Vesicular stomatitis Indiana virus/genetics , Animals , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Biomarkers, Tumor , Combined Modality Therapy , Disease Models, Animal , Gene Expression , Genetic Therapy/methods , Humans , Immunomodulation , Immunotherapy/methods , Interferon-beta/genetics , Interferon-beta/metabolism , Interferons/genetics , Interferons/metabolism , Melanoma, Experimental , Mice , Neoplasms/pathology , Neoplasms/therapy , Receptors, OX40/agonists , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transduction, Genetic , Transgenes , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Virus Replication , Xenograft Model Antitumor Assays
7.
Pediatr Blood Cancer ; 64(11)2017 Nov.
Article in English | MEDLINE | ID: mdl-28449314

ABSTRACT

Moxetumomab pasudotox is a second-generation recombinant immunotoxin against CD22 on B-cell lineages. Antileukemic activity has been demonstrated in children with chemotherapy-refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL), with variable responses. Here, we report in vitro and in vivo evaluation of moxetumomab pasudotox treatment of human cell lines and patient-derived cells as a preliminary study to understand characteristics of sensitivity to treatment. Binding, internalization, and apoptosis were evaluated using fluorescently tagged moxetumomab pasudotox. Studies in NOD-scid IL2Rgnull mice showed a modest survival benefit in mice engrafted with 697 cells but not in NALM6 or the two patient-derived xenograft models.


Subject(s)
Apoptosis/drug effects , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors , Adolescent , Adult , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Child , Child, Preschool , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult
8.
Am J Cancer Res ; 5(4): 1507-18, 2015.
Article in English | MEDLINE | ID: mdl-26101714

ABSTRACT

Variable clinical responses, tumor heterogeneity, and drug resistance reduce long-term survival outcomes for metastatic melanoma patients. To guide and accelerate drug development, we characterized tumor responses for five melanoma patient derived xenograft models treated with Vemurafenib. Three BRAF(V600E) models showed acquired drug resistance, one BRAF(V600E) model had a complete and durable response, and a BRAF(V600V) model was expectedly unresponsive. In progressing tumors, a variety of resistance mechanisms to BRAF inhibition were uncovered, including mutant BRAF alternative splicing, NRAS mutation, COT (MAP3K8) overexpression, and increased mutant BRAF gene amplification and copy number. The resistance mechanisms among the patient derived xenograft models were similar to the resistance pathways identified in clinical specimens from patients progressing on BRAF inhibitor therapy. In addition, there was both inter- and intra-patient heterogeneity in resistance mechanisms, accompanied by heterogeneous pERK expression immunostaining profiles. MEK monotherapy of Vemurafenib-resistant tumors caused toxicity and acquired drug resistance. However, tumors were eradicated when Vemurafenib was combined the MEK inhibitor. The diversity of drug responses among the xenograft models; the distinct mechanisms of resistance; and the ability to overcome resistance by the addition of a MEK inhibitor provide a scheduling rationale for clinical trials of next-generation drug combinations.

9.
J Transl Med ; 13: 110, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25889105

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is the most common type of solid bone cancer, with latent metastasis being a typical mode of disease progression and a major contributor to poor prognosis. For this to occur, cells must resist anoikis and be able to recapitulate tumorigenesis in a foreign microenvironment. Finding novel approaches to treat osteosarcoma and target those cell subpopulations that possess the ability to resist anoikis and contribute to metastatic disease is imperative. Here we investigate anchorage-independent (AI) cell growth as a model to better characterize anoikis resistance in human osteosarcoma while using an expression profiling approach to identify and test targetable signaling pathways. METHODS: Established human OS cell lines and patient-derived human OS cell isolates were subjected to growth in either adherent or AI conditions using Ultra-Low Attachment plates in identical media conditions. Growth rate was assessed using cell doubling times and chemoresistance was assessed by determining cell viability in response to a serial dilution of either doxorubicin or cisplatin. Gene expression differences were examined using quantitative reverse-transcription PCR and microarray with principal component and pathway analysis. In-vivo OS xenografts were generated by either subcutaneous or intratibial injection of adherent or AI human OS cells into athymic nude mice. Statistical significance was determined using student's t-tests with significance set at α=0.05. RESULTS: We show that AI growth results in a global gene expression profile change accompanied by significant chemoresistance (up to 75 fold, p<0.05). AI cells demonstrate alteration of key mediators of mesenchymal differentiation (ß-catenin, Runx2), stemness (Sox2), proliferation (c-myc, Akt), and epigenetic regulation (HDAC class 1). AI cells were equally tumorigenic as their adherent counterparts, but showed a significantly decreased rate of growth in-vitro and in-vivo (p<0.05). Treatment with the pan-histone deacetylase inhibitor vorinostat and the DNA methyltransferase inhibitor 5-azacytidine mitigated AI growth, while 5-azacytidine sensitized anoikis-resistant cells to doxorubicin (p<0.05). CONCLUSIONS: These data demonstrate remarkable plasticity in anoikis-resistant human osteosarcoma subpopulations accompanied by a rapid development of chemoresistance and altered growth rates mirroring the early stages of latent metastasis. Targeting epigenetic regulation of this process may be a viable therapeutic strategy.


Subject(s)
Anoikis , Bone Neoplasms/genetics , Epigenesis, Genetic , Gene Expression Profiling , Osteosarcoma/genetics , Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Osteosarcoma/drug therapy , Reverse Transcriptase Polymerase Chain Reaction
10.
Pediatr Blood Cancer ; 61(9): 1570-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24687871

ABSTRACT

BACKGROUND: Precision (Personalized) medicine has the potential to revolutionize patient health care especially for many cancers where the fundamental disease etiology remains either elusive or has no available therapy. Here we outline a study in alveolar rhabdomyosarcoma, in which we use gene expression profiling and a series of drug prediction algorithms combined with a matched patient-derived xenograft (PDX) model to test bioinformatically predicted therapies. PROCEDURE: A PDX model was developed from a patient biopsy and a number of drugs identified using gene expression analysis in combination with drug prediction algorithms. Drugs chosen from each of the predictive methodologies, along with the patient's standard-of-care therapy (ICE-T), were tested in vivo in the PDX tumor. A second study was initiated using the tumors that re-grew following the ICE-T treatment. Further expression analysis identified additional therapies with potential anti-tumor efficacy. RESULTS: A number of the predicted therapies were found to be active against the tumors in particular BGJ398 (FGFR2) and ICE-T. Re-transplanted ICE-T treated tumorgrafts demonstrated a decreased response to ICE-T recapitulating the patient's refractory disease. Gene expression profiling of the ICE-T treated tumorgrafts identified cytarabine (SLC29A1) as a potential therapy, which was shown, along with BGJ398, to be highly active in vivo. CONCLUSIONS: This study illustrates that PDX models are suitable surrogates for testing potential therapeutic strategies based on gene expression analysis, modeling clinical drug resistance and hold the potential to assist in guiding prospective patient care.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/genetics , Neoplasm Recurrence, Local/drug therapy , Precision Medicine , Rhabdomyosarcoma, Alveolar/drug therapy , Xenograft Model Antitumor Assays , Adult , Algorithms , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cytarabine/administration & dosage , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Oligonucleotide Array Sequence Analysis , Phenylurea Compounds/administration & dosage , Pyrimidines/administration & dosage , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/secondary
11.
J Transl Med ; 11: 213, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-24040940

ABSTRACT

BACKGROUND: Malignant peripheral nerve sheath tumors (MPNST) are rare highly aggressive sarcomas that affect 8-13% of people with neurofibromatosis type 1. The prognosis for patients with MPNST is very poor. Despite TOP2A overexpression in these tumors, doxorubicin resistance is common, and the mechanisms of chemotherapy resistance in MPNST are poorly understood. Molecular-guided therapy prediction is an emerging strategy for treatment refractory sarcomas that involves identification of therapy response and resistance mechanisms in individual tumors. Here, we report the results from a personalized, molecular-guided therapy analysis of MPNST samples. METHODS: Established molecular-guided therapy prediction software algorithms were used to analyze published microarray data from human MPNST samples and cell lines, with benign neurofibroma tissue controls. MPNST and benign neurofibroma-derived cell lines were used for confirmatory in vitro experimentation using quantitative real-time PCR and growth inhibition assays. Microarray data was analyzed using Affymetrix expression console MAS 5.0 method. Significance was calculated with Welch's t-test with non-corrected p-value < 0.05 and validated using permutation testing across samples. Paired Student's t-tests were used to compare relative EC50 values from independent growth inhibition experiments. RESULTS: Molecular guided therapy predictions highlight substantial variability amongst human MPNST samples in expression of drug target and drug resistance pathways, as well as some similarities amongst samples, including common up-regulation of DNA repair mechanisms. In a subset of MPNSTs, high expression of ABCC1 is observed, serving as a predicted contra-indication for doxorubicin and related therapeutics in these patients. These microarray-based results are confirmed with quantitative, real-time PCR and immunofluorescence. The functional effect of drug efflux in MPNST-derived cells is confirmed using in vitro growth inhibition assays. Alternative therapeutics supported by the molecular-guided therapy predictions are reported and tested in MPNST-derived cells. CONCLUSIONS: These results confirm the substantial molecular heterogeneity of MPNSTs and validate molecular-guided therapy predictions in vitro. The observed molecular heterogeneity in MPNSTs influences therapy prediction. Also, mechanisms involving drug transport and DNA damage repair are primary mediators of MPNST chemotherapy resistance. Together, these findings support the utility of individualized therapy in MPNST as in other sarcomas, and provide initial proof-of concept that individualized therapy prediction can be accomplished.


Subject(s)
Drug Resistance, Neoplasm , Molecular Targeted Therapy , Nerve Sheath Neoplasms/pathology , Nerve Sheath Neoplasms/therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Precision Medicine
12.
Lab Invest ; 93(8): 858-67, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23817084

ABSTRACT

Traditionally, the diagnosis of metastatic melanoma was terminal to most patients. However, the advancements towards understanding the fundamental etiology, pathophysiology, and treatment have raised melanoma to the forefront of contemporary medicine. Indeed, the evidence of durable remissions are being heard ever more frequently in clinics around the globe. Despite having more gene mutations per cell than any other type of cancer, investigators are overcoming complex genomic landscapes, signaling pathways, and immune checkpoints by generating novel technological methods and clinical protocols with breath-taking speed. Significant progress in deciphering molecular genetics, epigenetics, kinase-driven networks, metabolomics, and immune-enhancing pathways to achieve personalized and positive outcomes has truly provided new hope for melanoma patients. However, obstacles requiring breakthroughs include understanding the influence of sunlight exposure on melanoma etiology, and overcoming all too frequently acquired drug resistance, complicating targeted therapy. Pathologists continue to have critically important roles in advancing the field, particularly in the area of transitioning from microscope-based diagnostic reports to pharmacogenomics through molecularly informed tumor boards. Although melanoma is no longer considered just 'one disease', pathologists will continue this rapidly progressing and exciting journey to identify tumor subtypes, to utilize tumorgraft or so-called patient-derived xenograft (PDX) models, and to develop companion diagnostics to keep pace with the bewildering breakthroughs occurring on a regular basis. Exactly which combination of drugs will ultimately be required to eradicate melanoma cells remains to be determined. However, it is clear that pathologists who are as dedicated to melanoma as the pioneering pathologist Dr Sidney Farber was committed to childhood cancers, will be required as the battle against melanoma continues. In this review, we describe what sets melanoma apart from other tumors, and demonstrate how lessons learned in the melanoma clinic are being transferred to many other types of aggressive neoplasms.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Mutation , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Animals , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Genotype , Humans , Melanoma/drug therapy , Molecular Targeted Therapy , Phenotype , Precision Medicine , Remission Induction , Skin Neoplasms/drug therapy
13.
J Transl Med ; 11: 158, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23815880

ABSTRACT

BACKGROUND: A successful therapeutic strategy, specifically tailored to the molecular constitution of an individual and their disease, is an ambitious objective of modern medicine. In this report, we highlight a feasibility study in canine osteosarcoma focused on refining the infrastructure and processes required for prospective clinical trials using a series of gene expression-based Personalized Medicine (PMed) algorithms to predict suitable therapies within 5 days of sample receipt. METHODS: Tumor tissue samples were collected immediately following limb amputation and shipped overnight from veterinary practices. Upon receipt (day 1), RNA was extracted from snap-frozen tissue, with an adjacent H&E section for pathological diagnosis. Samples passing RNA and pathology QC were shipped to a CLIA-certified laboratory for genomic profiling. After mapping of canine probe sets to human genes and normalization against a (normal) reference set, gene level Z-scores were submitted to the PMed algorithms. The resulting PMed report was immediately forwarded to the veterinarians. Upon receipt and review of the PMed report, feedback from the practicing veterinarians was captured. RESULTS: 20 subjects were enrolled over a 5 month period. Tissue from 13 subjects passed both histological and RNA QC and were submitted for genomic analysis and subsequent PMed analysis and report generation. 11 of the 13 samples for which PMed reports were produced were communicated to the veterinarian within the target 5 business days. Of the 7 samples that failed QC, 4 were due to poor RNA quality, whereas 2 were failed following pathological review. Comments from the practicing veterinarians were generally positive and constructive, highlighting a number of areas for improvement, including enhanced education regarding PMed report interpretation, drug availability, affordable pricing and suitable canine dosing. CONCLUSIONS: This feasibility trial demonstrated that with the appropriate infrastructure and processes it is possible to perform an in-depth molecular analysis of a patient's tumor in support of real time therapeutic decision making within 5 days of sample receipt. A number of areas for improvement have been identified that should reduce the level of sample attrition and support clinical decision making.


Subject(s)
Dog Diseases/therapy , Osteosarcoma/veterinary , Precision Medicine , Animals , Dogs , Feasibility Studies , Female , Male , Osteosarcoma/therapy , Paraffin Embedding , Principal Component Analysis , Quality Control , Time Factors , Tissue Fixation
14.
PLoS One ; 8(4): e61088, 2013.
Article in English | MEDLINE | ID: mdl-23593398

ABSTRACT

Comparative oncology is a developing research discipline that is being used to assist our understanding of human neoplastic diseases. Companion canines are a preferred animal oncology model due to spontaneous tumor development and similarity to human disease at the pathophysiological level. We use a paired RNA sequencing (RNA-Seq)/microarray analysis of a set of four normal canine lymph nodes and ten canine lymphoma fine needle aspirates to identify technical biases and variation between the technologies and convergence on biological disease pathways. Surrogate Variable Analysis (SVA) provides a formal multivariate analysis of the combined RNA-Seq/microarray data set. Applying SVA to the data allows us to decompose variation into contributions associated with transcript abundance, differences between the technology, and latent variation within each technology. A substantial and highly statistically significant component of the variation reflects transcript abundance, and RNA-Seq appeared more sensitive for detection of transcripts expressed at low levels. Latent random variation among RNA-Seq samples is also distinct in character from that impacting microarray samples. In particular, we observed variation between RNA-Seq samples that reflects transcript GC content. Platform-independent variable decomposition without a priori knowledge of the sources of variation using SVA represents a generalizable method for accomplishing cross-platform data analysis. We identified genes differentially expressed between normal lymph nodes of disease free dogs and a subset of the diseased dogs diagnosed with B-cell lymphoma using each technology. There is statistically significant overlap between the RNA-Seq and microarray sets of differentially expressed genes. Analysis of overlapping genes in the context of biological systems suggests elevated expression and activity of PI3K signaling in B-cell lymphoma biopsies compared with normal biopsies, consistent with literature describing successful use of drugs targeting this pathway in lymphomas.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Animals , Base Composition , Cluster Analysis , Dogs , Genetic Variation , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Oligonucleotide Array Sequence Analysis , Signal Transduction
15.
J Transl Med ; 10: 125, 2012 Jun 18.
Article in English | MEDLINE | ID: mdl-22709571

ABSTRACT

BACKGROUND: There is resurgence within drug and biomarker development communities for the use of primary tumorgraft models as improved predictors of patient tumor response to novel therapeutic strategies. Despite perceived advantages over cell line derived xenograft models, there is limited data comparing the genotype and phenotype of tumorgrafts to the donor patient tumor, limiting the determination of molecular relevance of the tumorgraft model. This report directly compares the genomic characteristics of patient tumors and the derived tumorgraft models, including gene expression, and oncogenic mutation status. METHODS: Fresh tumor tissues from 182 cancer patients were implanted subcutaneously into immune-compromised mice for the development of primary patient tumorgraft models. Histological assessment was performed on both patient tumors and the resulting tumorgraft models. Somatic mutations in key oncogenes and gene expression levels of resulting tumorgrafts were compared to the matched patient tumors using the OncoCarta (Sequenom, San Diego, CA) and human gene microarray (Affymetrix, Santa Clara, CA) platforms respectively. The genomic stability of the established tumorgrafts was assessed across serial in vivo generations in a representative subset of models. The genomes of patient tumors that formed tumorgrafts were compared to those that did not to identify the possible molecular basis to successful engraftment or rejection. RESULTS: Fresh tumor tissues from 182 cancer patients were implanted into immune-compromised mice with forty-nine tumorgraft models that have been successfully established, exhibiting strong histological and genomic fidelity to the originating patient tumors. Comparison of the transcriptomes and oncogenic mutations between the tumorgrafts and the matched patient tumors were found to be stable across four tumorgraft generations. Not only did the various tumors retain the differentiation pattern, but supporting stromal elements were preserved. Those genes down-regulated specifically in tumorgrafts were enriched in biological pathways involved in host immune response, consistent with the immune deficiency status of the host. Patient tumors that successfully formed tumorgrafts were enriched for cell signaling, cell cycle, and cytoskeleton pathways and exhibited evidence of reduced immunogenicity. CONCLUSIONS: The preservation of the patient's tumor genomic profile and tumor microenvironment supports the view that primary patient tumorgrafts provide a relevant model to support the translation of new therapeutic strategies and personalized medicine approaches in oncology.


Subject(s)
Genomics , Neoplasms/genetics , Animals , Humans , Mice , Mice, Nude , Mutation , Neoplasms/pathology
16.
Cell Biol Int ; 34(9): 907-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20491673

ABSTRACT

This article discusses the need for transporter-mediated uptake for investigations addressing the mechanism of action of microcystins [with reference to the previous article in Cell Biology International by de Souza Votto (2007) 31:1359-1366].


Subject(s)
Membrane Transport Proteins/metabolism , Microcystins/metabolism , Models, Biological , Biological Transport/drug effects , Humans , Microcystins/toxicity , Organ Specificity/drug effects , Organic Anion Transporters/metabolism
17.
Toxicon ; 55(4): 827-37, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-19944114

ABSTRACT

Solute transporters that are selectively expressed on tumor cell membranes could be targeted with small molecule toxins that are selective substrates for these transporters. HeLa cells transfected to express the solute transporter OATP1B1 are exquisitely sensitive in vitro to microcystin LR (MCLR) and its analogs, and undergo rapid morphologic changes after exposure to MCLR. Immunoblot analyses revealed HSP27 phosphorylation increased prior to the rapid MCLR-induced morphologic changes. However, transfection of OATP1B1-expressing cells with HSP27 dominant negative mutants did not reverse MCLR toxicity. Although the MAP kinase p38 inhibitor SB202190 partially reversed MCLR cytotoxicity, the control molecule, SB202474, had similar effects. Unexpectedly, both SB202190 and SB202474 inhibited OATP1B1 uptake activity, indicating an alternative explanation for cytotoxicity reversal that did not involve p38 MAP kinase. Similarly, although the potassium chloride co-transporter (KCC) inhibitor (dihydro-indenyl)oxyalkanoic acid (DIOA), and the anti-oxidant, N-acetyl cysteine (NAC) both reversed MCLR cytotoxicity, both were also found to be unexpected OATP1B1 transport inhibitors. Therefore, the mechanism of MCLR-induced cytotoxicity is obscured by the inhibition of OATP1B1 uptake activity by MAP kinase inhibitors, DIOA, and NAC. Finally, growth of OATP1B1-expressing HeLa xenografts was inhibited by MCLR, suggesting that MCLR structural analogs selected for a broader therapeutic index could target OATP-expressing tumors.


Subject(s)
Acetylcysteine/pharmacology , Microcystins/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Organic Anion Transporters/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Blotting, Western , HeLa Cells , Humans , Liver-Specific Organic Anion Transporter 1 , Organic Anion Transporters/metabolism
18.
Cancer Chemother Pharmacol ; 66(1): 171-9, 2010 May.
Article in English | MEDLINE | ID: mdl-19830429

ABSTRACT

PURPOSE: We have previously shown that the expression of the thiamine transporter THTR2 is decreased sevenfold in breast cancer, which may leave breast cancer cells vulnerable to acute thiamine starvation. This concept was supported by the observation that MDA231 breast cancer xenografts demonstrated growth inhibition in mice fed a thiamine-free diet. METHODS: We purified recombinant Bacillus thiaminolyticus thiaminase I enzyme, which digests thiamine, to study acute thiamine starvation in breast cancer. RESULTS: Thiaminase I enzyme was cytotoxic in six breast cancer cell lines with IC(50)s ranging from 0.012 to 0.022 U/ml. The growth inhibitory effects of the combination of thiaminase I with either doxorubicin or paclitaxel were also examined. Over a wide range of drug concentrations, thiaminase 1 was consistently synergistic or additive with doxorubicin and paclitaxel in MCF-7, ZR75, HS578T and T47D cell lines, with most combinations having a calculated combination index (CI) of less than 0.8, indicating synergy. Although thiaminase I exposure did not stimulate the energy-sensing signaling kinases AKT, AMPK and GSK-3beta in MCF-7, ZR75, HS578T and T47D cell lines, thiaminase I exposure did stimulate expression of the ER stress response protein GRP78. In summary, thiaminase I is cytotoxic in breast cancer cell lines and triggers the unfolded protein response. CONCLUSION: These findings suggest that THTR2 down-regulation in breast tumors may present a nutritional vulnerability that could be exploited by thiaminase I enzyme therapy.


Subject(s)
Alkyl and Aryl Transferases/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Doxorubicin/therapeutic use , Paclitaxel/therapeutic use , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Alkyl and Aryl Transferases/administration & dosage , Animals , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/administration & dosage , Drug Screening Assays, Antitumor , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Heat-Shock Proteins/metabolism , Humans , Membrane Transport Proteins/metabolism , Mice , Mice, Nude , Paclitaxel/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Thiamine Deficiency/metabolism , Unfolded Protein Response/drug effects
19.
Mol Cancer Ther ; 6(2): 587-98, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17308056

ABSTRACT

Microcystins are a family of cyclic peptides that are potent inhibitors of the protein phosphatase families PP1 and PP2A. Only three human proteins are thought to be able to mediate the hepatic uptake of microcystins (the organic anion-transporting polypeptides OATP1B1, OATP1B3, and OATP1A2), and the predominant hepatic expression of these transporters accounts for the liver-specific toxicity of microcystins. A significant obstacle in the study of microcystins as anticancer drugs is the requirement of specific transport proteins for cellular uptake. We report that OATP1B3 mRNA is up-regulated in non-small cell lung cancer tumors in comparison with normal control tissues. This finding led to the exploration of microcystins as potential anticancer agents. We have developed a HeLa cell model with functional OATP1B1 and OATP1B3 activity. Transiently transfected HeLa cells are over 1,000-fold more sensitive to microcystin LR than the vector-transfected control cells, showing that transporter expression imparts marked selectivity for microcystin cytotoxicity. In addition, microcystin analogues showed variable cytotoxicities in the OATP1B1- and OATP1B3-transfected cells, including two analogues with IC(50) values <1 nmol/L. Cytotoxicity of microcystin analogues seems to correlate to the inhibition of PP2A in these cells and induces rapid cell death as seen by chromatin condensation and cell fragmentation. These studies show that microcystin-induced phosphatase inhibition results in potent cytotoxicity when microcystin compounds can gain intracellular access and are a potent novel class of therapeutic agents for tumors expressing these uptake proteins.


Subject(s)
Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Lung Neoplasms/metabolism , Microcystins/pharmacology , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/metabolism , Growth Inhibitors , HeLa Cells , Humans , Liver/metabolism , Liver-Specific Organic Anion Transporter 1 , Lung/metabolism , Lung Neoplasms/pathology , Marine Toxins , Microcystins/chemistry , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Peptides, Cyclic/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Solute Carrier Organic Anion Transporter Family Member 1B3 , Tumor Stem Cell Assay
20.
J Cell Biochem ; 98(1): 221-33, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16408291

ABSTRACT

Cancer cells in order to survive are often mutated to block apoptosis. One chemotherapeutic option is the re-establishment of apoptosis. An example of such a therapy is the PKC inhibitor Gö6976, which activates apoptosis and shrinks in vivo tumors in estrogen receptor-negative breast cancers. We proposed as a mechanism blockage of activation of the transcription factor NF-kappaB, which is anti-apoptotic and often elevated in cancers. Over recent years, questions have arisen regarding the specificity of these "small-molecule inhibitors." We have therefore explored the role of NF-kappaB inhibition in MDA-MB-231 breast cancer cells using small inhibitory RNAs (siRNA). siRNAs designed against NF-kappaB protein p65 (RelA) and IKKalpha, IKKbeta, and IKKgamma, strongly decreased the target proteins. But, unlike Gö6976, they did not decrease basal NF-kappaB or cause apoptosis. In particular, the decrease in p65 protein had no effects on apoptosis or cell proliferation, thus questioning the importance of NF-kappaB alone in the maintenance of these cells. Furthermore, the proteasome inhibitor MG-132 caused loss of IkappaBalpha, and an increase of it is phosphorylated form, but basal NF-kappaB was unchanged, whilst activation of NF-kappaB by TNFalpha was completely inhibited, suggesting that MG-132 activity is independent of constitutive NF-kappaB activation. We ascribe these differences to the specificity of inhibition by siRNAs as compared to the well-known non-specificity of small-molecule inhibitors. We conclude that the mutations in these cancer cells made them resistant to apoptosis, by elevating their NF-kappaB and activating other basal pathways that are blocked by Gö6976 but not by IKK and p65 siRNAs.


Subject(s)
Breast Neoplasms/metabolism , Gene Targeting , NF-kappa B/metabolism , RNA, Small Interfering/pharmacology , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Signal Transduction/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Targeting/methods , Humans , NF-kappa B/physiology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Receptors, Estrogen/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...