Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
Hum Vaccin Immunother ; 16(3): 636-644, 2020 03 03.
Article in English | MEDLINE | ID: mdl-31584324

ABSTRACT

Treatments with cytotoxic agents or viruses may cause Immunogenic Cell Death (ICD) that immunize tumor-bearing hosts but do not cause complete regression of tumor. We postulate that combining two ICD inducers may cause durable regression in immunocompetent mice. ICD was optimized in vitro by maximizing calreticulin externalization in human colorectal carcinoma (CRC) cells by exposure to mixtures of Oxaliplatin (OX) and human adenovirus (AdV). Six mm diameter CT26 or 4T1 carcinomas in flanks of BALB/c mice were injected once intratumorally (IT) with OX, AdV or their mixture. Tumor growth, Tumor-Infiltrating Lymphocytes (TIL), nodal cytotoxicity, and rejection of a viable cell challenge were measured. Tumors injected IT once with an optimum mixture of 80 µM OX - AdV 25 Multiplicity of Infection (MOI) in PBS buffer were 17-29% the volume of control tumors. When buffer was changed from PBS to 5% dextrose in water (D5W), volumes of tumors injected IT with 80 µM OX-AdV 25 MOI were 10% while IT OX or AdV alone were 32% and 40% the volume of IT buffer-treated tumors. OX-AdV IT increased CD3+ TIL by 4-fold, decreased CD8+ PD-1+ TIL from 79% to 19% and induced cytotoxicity to CT26 cells in draining node lymphocytes while lymphocytes from CT26-bearing untreated mice were not cytotoxic. OX-AdV IT in D5W caused complete regression in 40% of mice. Long-term survivors rejected a contralateral challenge of CT26. The buffer for Oxaliplatin is critical. The two ICD inducer mixture is promising as an agnostic sensitizer for carcinomas like colorectal carcinoma.


Subject(s)
Colorectal Neoplasms , Immunogenic Cell Death , Adenoviridae/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Oxaliplatin
2.
Mol Cancer Res ; 17(1): 42-53, 2019 01.
Article in English | MEDLINE | ID: mdl-30201826

ABSTRACT

MCL-1, a member of the antiapoptotic BCL-2 family, is a prosurvival protein with an essential DNA repair function. This study aims to test whether inhibition of protein synthesis by mTOR complex (mTORC) inhibitors depletes MCL-1, suppresses homologous recombination (HR) repair, and sensitizes cancer cells to PARP inhibitors. Treatment with everolimus decreases MCL-1 in colorectal carcinomas and small cell lung cancer (SCLC) cells but not glioblastoma multiforme (GBM) cells with a PTEN mutational background. However, AZD2014, a dual mTORC inhibitor, depletes MCL-1 in GBMs. Further, we show that everolimus decreases 4EBP1 phosphorylation only in colorectal carcinoma, whereas AZD2014 decreases 4EBP1 phosphorylation in both colorectal carcinoma and GBM cells. Combination therapy using everolimus or AZD2014 with olaparib inhibits the growth of clone A and U87-MG xenografts in in vivo and decreases clonogenic survival in in vitro compared with monotherapy. Reintroduction of MCL-1 rescues the survival of cancer cells in response to combination of everolimus or AZD2014 with olaparib. Treatment of cells with mTORC inhibitors and olaparib increases γ-H2AX and 53BP1 foci, decreases BRCA1, RPA, and Rad51 foci, impairs phosphorylation of ATR/Chk1 kinases, and induces necroptosis. In summary, mTORC inhibitors deplete MCL-1 to suppress HR repair and increase sensitivity to olaparib both in in vitro and in xenografts. IMPLICATIONS: Targeting the DNA repair activity of MCL-1 in in vivo for cancer therapy has not been tested. This study demonstrates that depleting MCL-1 sensitizes cancer cells to PARP inhibitors besides eliciting necroptosis, which could stimulate antitumor immunity to improve the therapeutic intervention of cancers.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/administration & dosage , Benzamides/pharmacology , Cell Line, Tumor , DNA Damage , Drug Repositioning , Drug Synergism , Everolimus/administration & dosage , Everolimus/pharmacology , Female , Humans , Male , Mice , Mice, Nude , Morpholines/administration & dosage , Morpholines/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasms/genetics , Neoplasms/metabolism , Phthalazines/administration & dosage , Phthalazines/pharmacology , Piperazines/administration & dosage , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
3.
Cytometry B Clin Cytom ; 94(2): 239-249, 2018 03.
Article in English | MEDLINE | ID: mdl-28475275

ABSTRACT

BACKGROUND: Minimal residual disease (MRD) in B lymphoblastic leukemia (B-ALL) by flow cytometry is an established prognostic factor used to adjust treatment in most pediatric therapeutic protocols. MRD in B-ALL has been standardized by the Children's Oncology Group (COG) in North America, but not routine clinical labs. The Foundation for National Institutes of Health sought to harmonize MRD measurement among COG, oncology groups, academic, community and government, laboratories. METHODS: Listmode data from post-induction marrows were distributed from a reference lab to seven different clinical FCM labs with variable experience in B-ALL MRD. Labs were provided with the COG protocol. Files from 15 cases were distributed to the seven labs. Educational sessions were implemented, and 10 more listmode file cases analyzed. RESULTS: Among 105 initial challenges, the overall discordance rate was 26%. In the final round, performance improved considerably; out of 70 challenges, there were five false positives and one false negative (9% discordance), and no quantitative discordance. Four of six deviations occurred in a single lab. Three samples with hematogones were still misclassified as MRD. CONCLUSIONS: Despite the provision of the COG standardized analysis protocol, even experienced laboratories require an educational component for B-ALL MRD analysis by FCM. Recognition of hematogones remains challenging for some labs when using the COG protocol. The results from this study suggest that dissemination of MRD testing to other North American laboratories as part of routine clinical management of B-ALL is possible but requires additional educational components to complement standardized methodology. © 2017 International Clinical Cytometry Society.


Subject(s)
Neoplasm, Residual/diagnosis , Neoplasm, Residual/pathology , Flow Cytometry/methods , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis
4.
JAMA Oncol ; 3(7): e170580, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28494052

ABSTRACT

IMPORTANCE: Minimal residual disease (MRD) refers to the presence of disease in cases deemed to be in complete remission by conventional pathologic analysis. Assessing the association of MRD status following induction therapy in patients with acute lymphoblastic leukemia (ALL) with relapse and mortality may improve the efficiency of clinical trials and accelerate drug development. OBJECTIVE: To quantify the relationships between event-free survival (EFS) and overall survival (OS) with MRD status in pediatric and adult ALL using publications of clinical trials and other databases. DATA SOURCES: Clinical studies in ALL identified via searches of PubMed, MEDLINE, and clinicaltrials.gov. STUDY SELECTION: Our search and study screening process adhered to the PRISMA Guidelines. Studies that addressed EFS or OS by MRD status in patients with ALL were included; reviews, abstracts, and studies with fewer than 30 patients or insufficient MRD description were excluded. DATA EXTRACTION AND SYNTHESIS: Study sample size, patient age, follow-up time, timing of MRD assessment (postinduction or consolidation), MRD detection method, phenotype/genotype (B cell, T cell, Philadelphia chromosome), and EFS and OS. Searches of PubMed and MEDLINE identified 566 articles. A parallel search on clinicaltrials.gov found 67 closed trials and 62 open trials as of 2014. Merging results of 2 independent searches and applying exclusions gave 39 publications in 3 arms of patient populations (adult, pediatric, and mixed). We performed separate meta-analyses for each of these 3 subpopulations. RESULTS: The 39 publications comprised 13 637 patients: 16 adult studies (2076 patients), 20 pediatric (11 249 patients), and 3 mixed (312 patients). The EFS hazard ratio (HR) for achieving MRD negativity is 0.23 (95% Bayesian credible interval [BCI] 0.18-0.28) for pediatric patients and 0.28 (95% BCI, 0.24-0.33) for adults. The respective HRs in OS are 0.28 (95% BCI, 0.19-0.41) and 0.28 (95% BCI, 0.20-0.39). The effect was similar across all subgroups and covariates. CONCLUSIONS AND RELEVANCE: The value of having achieved MRD negativity is substantial in both pediatric and adult patients with ALL. These results are consistent across therapies, methods of and times of MRD assessment, cutoff levels, and disease subtypes. Minimal residual disease status warrants consideration as an early measure of disease response for evaluating new therapies, improving the efficiency of clinical trials, accelerating drug development, and for regulatory approval. A caveat is that an accelerated approval of a particular new drug using an intermediate end point, such as MRD, would require confirmation using traditional efficacy end points.


Subject(s)
Neoplasm, Residual/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adult , Child , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Prognosis , Survival Analysis , Treatment Outcome
5.
Ann Oncol ; 28(5): 1023-1031, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28453697

ABSTRACT

Background: TNM staging alone does not accurately predict outcome in colon cancer (CC) patients who may be eligible for adjuvant chemotherapy. It is unknown to what extent the molecular markers microsatellite instability (MSI) and mutations in BRAF or KRAS improve prognostic estimation in multivariable models that include detailed clinicopathological annotation. Patients and methods: After imputation of missing at random data, a subset of patients accrued in phase 3 trials with adjuvant chemotherapy (n = 3016)-N0147 (NCT00079274) and PETACC3 (NCT00026273)-was aggregated to construct multivariable Cox models for 5-year overall survival that were subsequently validated internally in the remaining clinical trial samples (n = 1499), and also externally in different population cohorts of chemotherapy-treated (n = 949) or -untreated (n = 1080) CC patients, and an additional series without treatment annotation (n = 782). Results: TNM staging, MSI and BRAFV600E mutation status remained independent prognostic factors in multivariable models across clinical trials cohorts and observational studies. Concordance indices increased from 0.61-0.68 in the TNM alone model to 0.63-0.71 in models with added molecular markers, 0.65-0.73 with clinicopathological features and 0.66-0.74 with all covariates. In validation cohorts with complete annotation, the integrated time-dependent AUC rose from 0.64 for the TNM alone model to 0.67 for models that included clinicopathological features, with or without molecular markers. In patient cohorts that received adjuvant chemotherapy, the relative proportion of variance explained (R2) by TNM, clinicopathological features and molecular markers was on an average 65%, 25% and 10%, respectively. Conclusions: Incorporation of MSI, BRAFV600E and KRAS mutation status to overall survival models with TNM staging improves the ability to precisely prognosticate in stage II and III CC patients, but only modestly increases prediction accuracy in multivariable models that include clinicopathological features, particularly in chemotherapy-treated patients.


Subject(s)
Biomarkers, Tumor/metabolism , Colonic Neoplasms/mortality , Adult , Aged , Aged, 80 and over , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Proportional Hazards Models , Retrospective Studies , Treatment Outcome , Young Adult
6.
Clin Cancer Res ; 23(15): 3980-3993, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28428191

ABSTRACT

Treatment of myeloma has benefited from the introduction of more effective and better tolerated agents, improvements in supportive care, better understanding of disease biology, revision of diagnostic criteria, and new sensitive and specific tools for disease prognostication and management. Assessment of minimal residual disease (MRD) in response to therapy is one of these tools, as longer progression-free survival (PFS) is seen consistently among patients who have achieved MRD negativity. Current therapies lead to unprecedented frequency and depth of response, and next-generation flow and sequencing methods to measure MRD in bone marrow are in use and being developed with sensitivities in the range of 10-5 to 10-6 cells. These technologies may be combined with functional imaging to detect MRD outside of bone marrow. Moreover, immune profiling methods are being developed to better understand the immune environment in myeloma and response to immunomodulatory agents while methods for molecular profiling of myeloma cells and circulating DNA in blood are also emerging. With the continued development and standardization of these methodologies, MRD has high potential for use in gaining new drug approvals in myeloma. The FDA has outlined two pathways by which MRD could be qualified as a surrogate endpoint for clinical studies directed at obtaining accelerated approval for new myeloma drugs. Most importantly, better understanding of MRD should also contribute to better treatment monitoring. Potentially, MRD status could be used as a prognostic factor for making treatment decisions and for informing timing of therapeutic interventions. Clin Cancer Res; 23(15); 3980-93. ©2017 AACR.


Subject(s)
Circulating Tumor DNA/blood , Multiple Myeloma/blood , Multiple Myeloma/drug therapy , Neoplasm, Residual/blood , Biomarkers, Tumor/genetics , Bone Marrow/drug effects , Bone Marrow/pathology , Disease-Free Survival , High-Throughput Nucleotide Sequencing/methods , Humans , Multiple Myeloma/complications , Multiple Myeloma/genetics , Neoplasm, Residual/chemically induced , Neoplasm, Residual/genetics , Patient Selection , Prognosis
7.
Oper Dent ; 42(3): 266-272, 2017.
Article in English | MEDLINE | ID: mdl-28080293

ABSTRACT

The carving of a complex amalgam restoration may occasionally result in light proximal contact with the adjacent tooth. The purpose of this study was to investigate the strength of complex amalgam restorations repaired with a proximal slot amalgam preparation. Extracted human third molars of similar coronal size were sectioned 1 mm apical to the height of the contour using a saw and were randomly distributed into 9 groups of 10 teeth each. One pin was placed at each line angle of the flattened dentinal tooth surface. A metal matrix band was placed and an admixed alloy was condensed and carved to create a full crown contour but with a flat occlusal surface. A proximal slot was prepared with or without a retention groove and repaired using a single-composition spherical amalgam 15 minutes, 24 hours, one week, or six months after the initial crown condensation. The specimens were stored for 24 hours in 37°C water before fracture at the marginal ridge using a round-ended blade in a universal testing machine. The control group was not repaired. The mean maximum force in newtons and standard deviation were determined per group. Data were analyzed with a 2-way analysis of variance as well as Tukey and Dunnett tests (α=0.05). Significant differences were found between groups based on type of slot preparation (p=0.017) but not on time (p=0.327), with no significant interaction (p=0.152). No significant difference in the strength of the marginal ridge was found between any repair group and the unrepaired control group (p>0.076). The proximal repair strength of a complex amalgam restoration was not significantly different from an unrepaired amalgam crown. Placing a retention groove in the proximal slot preparation resulted in significantly greater fracture strength than a slot with no retention grooves. Time of repair had no significant effect on the strength of the repair.


Subject(s)
Dental Amalgam/chemistry , Dental Cavity Preparation/methods , Dental Restoration, Permanent/methods , Dental Materials/chemistry , Dental Restoration Failure , Dental Stress Analysis , Humans , In Vitro Techniques , Materials Testing , Molar, Third , Time Factors
8.
Am J Gastroenterol ; 110(7): 948-55, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25601013

ABSTRACT

OBJECTIVES: As there are no US population-based studies examining Lynch syndrome (LS) screening frequency by microsatellite instability (MSI) and immunohistochemistry (IHC), we seek to quantitate statewide rates in patients aged ≤50 years using data from a Centers for Disease Control and Prevention-funded Comparative Effectiveness Research (CER) project and identify factors associated with testing. Screening rates in this young, high-risk population may provide a best-case scenario as older patients, potentially deemed lower risk, may undergo testing less frequently. We also seek to determine how frequently MSI/IHC results are available preoperatively, as this may assist with decisions regarding colonic resection extent. METHODS: Data from all Louisiana colorectal cancer (CRC) patients aged ≤50 years diagnosed in 2011 were obtained from the Louisiana Tumor Registry CER project. Registry researchers and physicians analyzed data, including pathology and MSI/IHC. RESULTS: Of the 2,427 statewide all-age CRC patients, there were 274 patients aged ≤50 years, representing health care at 61 distinct facilities. MSI and/or IHC were performed in 23.0% of patients. Testing-associated factors included CRC family history (P<0.0045), urban location (P<0.0370), and care at comprehensive cancer centers (P<0.0020) but not synchronous/metachronous CRC or MSI-like histology. Public hospital screening was disproportionately low (P<0.0217). Of those tested, MSI and/or IHC was abnormal in 21.7%. Of those with abnormal IHC, staining patterns were consistent with LS in 87.5%. MSI/IHC results were available preoperatively in 16.9% of cases. CONCLUSIONS: Despite frequently abnormal MSI/IHC results, LS screening in young, high-risk patients is low. Provider education and disparities in access to specialized services, particularly in underserved populations, are possible contributors. MSI/IHC results are infrequently available preoperatively.


Subject(s)
Colectomy , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Early Detection of Cancer , Genetic Testing , Mass Screening , Microsatellite Instability , Adult , Age Factors , Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , Colorectal Neoplasms, Hereditary Nonpolyposis/prevention & control , Colorectal Neoplasms, Hereditary Nonpolyposis/surgery , Confounding Factors, Epidemiologic , Early Detection of Cancer/methods , Early Detection of Cancer/standards , Early Detection of Cancer/statistics & numerical data , Female , Humans , Immunohistochemistry , Louisiana/epidemiology , Male , Mass Screening/methods , Mass Screening/standards , Mass Screening/statistics & numerical data , Middle Aged , Neoplasms, Multiple Primary/genetics , Neoplasms, Second Primary/genetics , Preoperative Period , Prevalence , Rural Population/statistics & numerical data , Time Factors , United States/epidemiology , Urban Population/statistics & numerical data
9.
Cancer ; 120 Suppl 23: 3793-806, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25412391

ABSTRACT

BACKGROUND: The Collaborative Stage (CS) Data Collection System enables multiple cancer registration programs to document anatomic and molecular pathology features that contribute to the Tumor (T), Node (N), Metastasis (M) - TNM - system of the American Joint Committee on Cancer (AJCC). This article highlights changes in CS for colon and rectal carcinomas as TNM moved from the AJCC 6th to the 7th editions. METHODS: Data from 18 Surveillance, Epidemiology, and End Results (SEER) population-based registries were analyzed for the years 2004-2010, which included 191,361colon and 73,341 rectal carcinomas. RESULTS: Overall, the incidence of colon and rectal cancers declined, with the greatest decrease in stage 0. The AJCC's 7th edition introduction of changes in the subcategorization of T4, N1, and N2 caused shifting within stage groups in 25,577 colon and 10,150 rectal cancers diagnosed in 2010. Several site-specific factors (SSFs) introduced in the 7th edition had interesting findings: 1) approximately 10% of colon and rectal cancers had tumor deposits - about 30%-40% occurred without lymph node metastases, which resulted in 2.5% of colon and 3.3% of rectal cases becoming N1c (stage III A/B) in the AJCC 7th edition; 2) 10% of colon and 12% of rectal cases had circumferential radial margins <1 mm; 3) about 46% of colorectal cases did not have a carcinoembryonic antigen (CEA) testing or documented CEA information; and 4) about 10% of colorectal cases had perineural invasion. CONCLUSIONS: Adoption of the AJCC 7th edition by the SEER program provides an assessment tool for staging and SSFs on clinical outcomes. This evidence can be used for education and improved treatment for colorectal carcinomas.


Subject(s)
Carcinoma/pathology , Colonic Neoplasms/pathology , Lymph Nodes/pathology , Rectal Neoplasms/pathology , Registries , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoembryonic Antigen/blood , Carcinoma/metabolism , Cohort Studies , Colonic Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Neoplasm Staging/trends , Rectal Neoplasms/metabolism , Retrospective Studies , SEER Program
10.
Clin Cancer Res ; 20(21): 5446-55, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25208882

ABSTRACT

PURPOSE: High levels of BCL-2 family members in colorectal carcinoma cause resistance to treatment. Inhibition of NANOG or its paralog NANOGP8 reduces the proliferation, stemness, and tumorigenicity of colorectal carcinoma cells. Our hypothesis was that inhibition of NANOG/NANOGP8 enhances the cytotoxic effect of BH3 mimetics targeting BCL-2 family members in colorectal carcinoma cells through reducing expression of MCL-1, a prosurvival BCL-2 protein. EXPERIMENTAL DESIGN: Lentiviral vector (LV) shRNA to NANOG (shNG-1) or NANOGP8 (shNp8-1) transduced colorectal carcinoma cells that were also exposed to the BH3 mimetics ABT-737 or ABT-199 in vivo in colorectal carcinoma xenografts and in vitro where proliferation, protein and gene expression, and apoptosis were measured. RESULTS: Clone A and CX-1 were sensitive to ABT-737 and ABT-199 at IC50s of 2 to 9 µmol/L but LS174T was resistant with IC50s of 18 to 30 µmol/L. Resistance was associated with high MCL-1 expression in LS174T. LVshNG-1 or LVshNp8-1 decreased MCL-1 expression, increased apoptosis, and decreased replating efficiency in colorectal carcinoma cells treated with either ABT-737 or ABT-199 compared with the effects of either BH3 mimetic alone. Inhibition or overexpression of MCL-1 alone replicated the effects of LVshNG-1 or LVshNp8-1 in increasing or decreasing the apoptosis caused with the BH3 mimetic. The combination therapy inhibited the growth of LS174T xenografts in vivo compared with untreated controls or treatment with only LV shRNA or ABT-737. CONCLUSIONS: Inhibition of NANOGP8 or NANOG enhances the cytotoxicity of BH3 mimetics that target BCL-2 family members. Gene therapy targeting the NANOGs may increase the efficacy of BH3 mimetics in colorectal carcinoma.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/genetics , Colorectal Neoplasms/genetics , Down-Regulation/genetics , Homeodomain Proteins/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biphenyl Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Down-Regulation/drug effects , HEK293 Cells , HT29 Cells , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Nanog Homeobox Protein , Nitrophenols/pharmacology , Piperazines/pharmacology , RNA, Small Interfering/genetics , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 20(6): 1428-44, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24634466

ABSTRACT

This article defines and describes best practices for the academic and business community to generate evidence of clinical utility for cancer molecular diagnostic assays. Beyond analytical and clinical validation, successful demonstration of clinical utility involves developing sufficient evidence to demonstrate that a diagnostic test results in an improvement in patient outcomes. This discussion is complementary to theoretical frameworks described in previously published guidance and literature reports by the U.S. Food and Drug Administration, Centers for Disease Control and Prevention, Institute of Medicine, and Center for Medical Technology Policy, among others. These reports are comprehensive and specifically clarify appropriate clinical use, adoption, and payer reimbursement for assay manufacturers, as well as Clinical Laboratory Improvement Amendments-certified laboratories, including those that develop assays (laboratory developed tests). Practical criteria and steps for establishing clinical utility are crucial to subsequent decisions for reimbursement without which high-performing molecular diagnostics will have limited availability to patients with cancer and fail to translate scientific advances into high-quality and cost-effective cancer care. See all articles in this CCR Focus section, "The Precision Medicine Conundrum: Approaches to Companion Diagnostic Co-development."


Subject(s)
Diagnostic Test Approval , Molecular Diagnostic Techniques , Neoplasms/diagnosis , Practice Guidelines as Topic , Diagnostic Test Approval/standards , Diagnostic Test Approval/trends , Humans , Molecular Diagnostic Techniques/standards , Molecular Diagnostic Techniques/trends , Molecular Targeted Therapy/methods , Neoplasms/therapy , Practice Guidelines as Topic/standards , United States
12.
Oncogene ; 32(37): 4397-405, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23085761

ABSTRACT

NANOG is a stem cell transcription factor that is essential for embryonic development, reprogramming normal adult cells and malignant transformation and progression. The nearly identical retrogene NANOGP8 is expressed in multiple cancers, but generally not in normal tissues and its function is not well defined. Our postulate is that NANOGP8 directly modulates the stemness of individual human colorectal carcinoma (CRC) cells. Stemness was measured in vitro as the spherogenicity of single CRC cells in serum-free medium and the size of the side population (SP) and in vivo as tumorigenicity and experimental metastatic potential in NOD/SCID mice. We found that 80% of clinical liver metastases express a NANOG with 75% of the positive metastases containing NANOGP8 transcripts. In all, 3-62% of single cells within six CRC lines form spheroids in serum-free medium in suspension. NANOGP8 is translated into protein. The relative expression of a NANOG gene increased 8- to 122-fold during spheroid formation, more than the increase in OCT4 or SOX2 transcripts with NANOGP8 the more prevalent family member. Short hairpin RNA (shRNA) to NANOG not only inhibits spherogenicity but also reduces expression of OCT4 and SOX2, the size of the SP and tumor growth in vivo. Inhibition of NANOG gene expression is associated with inhibition of proliferation and decreased phosphorylation of G2-related cell-cycle proteins. Overexpression of NANOGP8 rescues single-cell spherogenicity when NANOG gene expression is inhibited and increases the SP in CRC. Thus, NANOGP8 can substitute for NANOG in directly promoting stemness in CRC.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nanog Homeobox Protein , Neoplasm Metastasis , Spheroids, Cellular , Tumor Cells, Cultured
13.
J Clin Oncol ; 30(19): 2327-33, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22585691

ABSTRACT

PURPOSE: Surgical resection of gastric cancer has produced suboptimal survival despite multiple randomized trials that used postoperative chemotherapy or more aggressive surgical procedures. We performed a randomized phase III trial of postoperative radiochemotherapy in those at moderate risk of locoregional failure (LRF) following surgery. We originally reported results with 4-year median follow-up. This update, with a more than 10-year median follow-up, presents data on failure patterns and second malignancies and explores selected subset analyses. PATIENTS AND METHODS: In all, 559 patients with primaries ≥ T3 and/or node-positive gastric cancer were randomly assigned to observation versus radiochemotherapy after R0 resection. Fluorouracil and leucovorin were administered before, during, and after radiotherapy. Radiotherapy was given to all LRF sites to a dose of 45 Gy. RESULTS: Overall survival (OS) and relapse-free survival (RFS) data demonstrate continued strong benefit from postoperative radiochemotherapy. The hazard ratio (HR) for OS is 1.32 (95% CI, 1.10 to 1.60; P = .0046). The HR for RFS is 1.51 (95% CI, 1.25 to 1.83; P < .001). Adjuvant radiochemotherapy produced substantial reduction in both overall relapse and locoregional relapse. Second malignancies were observed in 21 patients with radiotherapy versus eight with observation (P = .21). Subset analyses show robust treatment benefit in most subsets, with the exception of patients with diffuse histology who exhibited minimal nonsignificant treatment effect. CONCLUSION: Intergroup 0116 (INT-0116) demonstrates strong persistent benefit from adjuvant radiochemotherapy. Toxicities, including second malignancies, appear acceptable, given the magnitude of RFS and OS improvement. LRF reduction may account for the majority of overall relapse reduction. Adjuvant radiochemotherapy remains a rational standard therapy for curatively resected gastric cancer with primaries T3 or greater and/or positive nodes.


Subject(s)
Adenocarcinoma/therapy , Gastrectomy/methods , Stomach Neoplasms/surgery , Stomach Neoplasms/therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Adenocarcinoma/surgery , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy, Adjuvant , Female , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Male , Stomach Neoplasms/drug therapy , Stomach Neoplasms/radiotherapy , Survival Analysis , Treatment Outcome
14.
Clin Cancer Res ; 18(6): 1515-23, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22422403

ABSTRACT

Molecular diagnostics are becoming increasingly important in clinical research to stratify or identify molecularly profiled patient cohorts for targeted therapies, to modify the dose of a therapeutic, and to assess early response to therapy or monitor patients. Molecular diagnostics can also be used to identify the pharmacogenetic risk of adverse drug reactions. The articles in this CCR Focus section on molecular diagnosis describe the development and use of markers to guide medical decisions regarding cancer patients. They define sources of preanalytic variability that need to be minimized, as well as the regulatory and financial challenges involved in developing diagnostics and integrating them into clinical practice. They also outline a National Cancer Institute program to assist diagnostic development. Molecular diagnostic clinical tests require rigor in their development and clinical validation, with sensitivity, specificity, and validity comparable to those required for the development of therapeutics. These diagnostics must be offered at a realistic cost that reflects both their clinical value and the costs associated with their development. When genome-sequencing technologies move into the clinic, they must be integrated with and traceable to current technology because they may identify more efficient and accurate approaches to drug development. In addition, regulators may define progressive drug approval for companion diagnostics that requires further evidence regarding efficacy and safety before full approval can be achieved. One way to accomplish this is to emphasize phase IV postmarketing, hypothesis-driven clinical trials with biological characterization that would permit an accurate definition of the association of low-prevalence gene alterations with toxicity or response in large cohorts.


Subject(s)
Biomarkers, Tumor , Drug Design , Neoplasms/diagnosis , Pathology, Molecular/standards , Biological Specimen Banks , Clinical Trials as Topic , Databases as Topic , Humans , Molecular Diagnostic Techniques , Neoplasms/drug therapy , Neoplasms/genetics , Validation Studies as Topic
15.
Clin Cancer Res ; 18(6): 1531-9, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22422405

ABSTRACT

The development of clinically useful molecular diagnostics requires validation of clinical assay performance and achievement of clinical qualification in clinical trials. As discussed elsewhere in this Focus section on molecular diagnostics, validation of assay performance must be rigorous, especially when the assay will be used to guide treatment decisions. Here we review some of the problems associated with assay development, especially for academic investigators. These include lack of expertise and resources for analytical validation, lack of experience in designing projects for a specific clinical use, lack of specimens from appropriate patient groups, and lack of access to Clinical Laboratory Improvement Amendments-certified laboratories. In addition, financial support for assay validation has lagged behind financial support for marker discovery or drug development, even though the molecular diagnostic may be considered necessary for the successful use of the companion therapeutic. The National Cancer Institute supports a large number of clinical trials and a significant effort in drug development. In order to address some of these barriers for predictive and prognostic assays that will be used in clinical trials to select patients for a particular treatment, stratify patients into molecularly defined subgroups, or choose between treatments for molecularly defined tumors, the National Cancer Institute has begun a pilot program designed to lessen barriers to the development of validated prognostic and predictive assays.


Subject(s)
Biomarkers, Tumor/analysis , Pathology, Molecular/standards , Biomedical Research , Clinical Trials as Topic , Humans , National Institutes of Health (U.S.) , Neoplasms/diagnosis , Pilot Projects , Prognosis , United States , Validation Studies as Topic
16.
Clin Cancer Res ; 18(6): 1547-54, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22422407

ABSTRACT

The U.S. Food and Drug Administration is now exerting its regulatory authority over the use of molecular diagnostics and related assays for medical decision making in clinical trials, by performing pre-Investigational Device Exemption reviews in all phases of clinical trials. In this review, we assess the analytical performance of the assay for the diagnostic, and consider how that performance affects the diagnostic and the patient and their risks and benefits from treatment. We also discuss the process involved in the first review of a new Children's Oncology Group phase III trial in acute myelogenous leukemia. The lessons learned and recommendations for how to prepare for and incorporate this new level of regulatory review into the protocol development process are presented.


Subject(s)
Device Approval , Government Regulation , Molecular Diagnostic Techniques , United States Food and Drug Administration , Biomarkers, Tumor/analysis , Clinical Trials, Phase III as Topic , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Randomized Controlled Trials as Topic , United States , Validation Studies as Topic
17.
PLoS One ; 7(2): e31058, 2012.
Article in English | MEDLINE | ID: mdl-22347427

ABSTRACT

Exposure to oxygen-rich environments can lead to oxidative damage, increased body iron stores, and changes in status of some vitamins, including folate. Assessing the type of oxidative damage in these environments and determining its relationships with changes in folate status are important for defining nutrient requirements and designing countermeasures to mitigate these effects. Responses of humans to oxidative stressors were examined in participants undergoing a saturation dive in an environment with increased partial pressure of oxygen, a NASA Extreme Environment Mission Operations mission. Six participants completed a 13-d saturation dive in a habitat 19 m below the ocean surface near Key Largo, FL. Fasting blood samples were collected before, twice during, and twice after the dive and analyzed for biochemical markers of iron status, oxidative damage, and vitamin status. Body iron stores and ferritin increased during the dive (P<0.001), with a concomitant decrease in RBC folate (P<0.001) and superoxide dismutase activity (P<0.001). Folate status was correlated with serum ferritin (Pearson r = -0.34, P<0.05). Peripheral blood mononuclear cell poly(ADP-ribose) increased during the dive and the increase was significant by the end of the dive (P<0.001); γ-H2AX did not change during the mission. Together, the data provide evidence that when body iron stores were elevated in a hyperoxic environment, a DNA damage repair response occurred in peripheral blood mononuclear cells, but double-stranded DNA damage did not. In addition, folate status decreases quickly in this environment, and this study provides evidence that folate requirements may be greater when body iron stores and DNA damage repair responses are elevated.


Subject(s)
DNA Damage , DNA Repair , Diving/adverse effects , Folic Acid/blood , Biomarkers/blood , Blood Chemical Analysis , Humans , Hyperoxia , Iron/analysis , Oxygen , United States , United States National Aeronautics and Space Administration
18.
J Pathol ; 221(4): 361-2, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20593484

ABSTRACT

This Invited Response addresses concerns and opinions expressed in an Invited Commentary, 'Evidence-based medicine: the time has come to set standards for staging', by Quirke et al., published in this issue of The Journal of Pathology.


Subject(s)
Neoplasm Staging/standards , Neoplasms/pathology , Colorectal Neoplasms/pathology , Evidence-Based Medicine/methods , Humans , Lung Neoplasms/pathology , Lymphatic Metastasis , Neoplasm Staging/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...