Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
2.
J Gastrointest Cancer ; 54(4): 1017-1030, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37009977

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors have recently replaced over chemotherapy as the first-line treatment for microsatellite instability-high or mismatch repair deficient (dMMR/MSI-H) stage 4 colorectal cancers. Considering this success, many studies have tried to replicate the use of immune checkpoint inhibitors, either as a single agent or in combination with other therapeutic agents, in the treatment of proficient mismatch repair (pMMR/MSS) stage 4 colorectal cancers. This review summarizes the seminal clinical data about the immune checkpoint inhibitors used in pMMR/MSS colorectal cancers and some future directions. RESULTS: Studies concerning the use of immune checkpoint inhibitors as a single agent or in combination with other immune checkpoint inhibitors, targeted therapy, chemotherapy, or radiotherapy have proven inefficient in the treatment of pMMR/MSS colorectal cancer. However, a small subset of patients with pMMR/MSS colorectal cancer who has a mutation in POLE and POLD1 enzymes may respond to immunotherapy. Moreover, patients without liver metastasis appear to have a better chance of response. New immune checkpoint targets are being identified, such as VISTA, TIGIT, LAG3, STING signal pathway, and BTLA, and studies are ongoing to determine their efficiency in this disease type. CONCLUSION: Immune checkpoint inhibitor-based regimens have not yet shown any meaningful positive outcomes for most pMMR/MSS colorectal cancers. A beneficial effect among a minority of these patients has been observed, but concrete biomarkers of response are lacking. Understanding the underlying mechanisms of immune resistance should guide further research for overcoming these obstacles.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Humans , DNA Mismatch Repair , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Microsatellite Instability
3.
Clin Colorectal Cancer ; 21(4): 315-324, 2022 12.
Article in English | MEDLINE | ID: mdl-36283915

ABSTRACT

BACKGROUND: Adjuvant chemotherapy for stage III colon cancer is underutilized in the United States. The aim of this study was to assess the use of adjuvant chemotherapy in younger and medically fit patients and analyze the socioeconomic factors associated with its utilization. METHODS: Using the National Cancer Database from 2004 to 2015, we selected stage III colon cancer patients between age 18 to 65, Charlson-Deyo Comorbidity Index (CDCI) of 0 or 1, and those that survived at least 12 months after surgery. We then compared patients that underwent surgery only with those that received adjuvant chemotherapy. Multivariable logistic regression analysis was performed to identify variables associated with adjuvant chemotherapy use in the population. Overall survival was estimated by Kaplan-Meier curves. RESULTS: Of the 48,336 patients that met inclusion criteria, 43,315 (90%) received adjuvant chemotherapy. The utilization of adjuvant chemotherapy increased from 87% in 2004 to 91% in 2015. On multivariable regression analysis, the use of adjuvant chemotherapy was lower among males, Non-Hispanic Blacks and Hispanics, low-grade cancer, left-sided tumors, CDCI 1, those who travel ≥ 50 miles, yearly income < $40,227, and uninsured patients. The most common reason for the omission of adjuvant chemotherapy was the patient or caregiver's choice (40% between 2013 and 2015). The 5-year and 10-year overall survival rates were 76.7% and 63.8% respectively, in those who received adjuvant chemotherapy as compared to 65.1% and 49.3% in those who underwent surgery only (P < .001). CONCLUSION: In young and medically fit stage III colon cancer patients, most patients received guideline-compliant care in the United States. However, socioeconomic disparities adversely impacted the use of adjuvant chemotherapy. The patient or caregiver's decision was the most common reason for non-adherence to adjuvant chemotherapy and lead to poor survival outcomes. Emphasis should be placed on developing patient-centered strategies to improve adherence to chemotherapy in all patients.


Subject(s)
Colonic Neoplasms , Male , Humans , United States/epidemiology , Middle Aged , Adolescent , Young Adult , Adult , Aged , Neoplasm Staging , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Colonic Neoplasms/epidemiology , Comorbidity , Survival Rate
4.
Cell ; 185(2): 283-298.e17, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35021065

ABSTRACT

Gasdermins are a family of structurally related proteins originally described for their role in pyroptosis. Gasdermin B (GSDMB) is currently the least studied, and while its association with genetic susceptibility to chronic mucosal inflammatory disorders is well established, little is known about its functional relevance during active disease states. Herein, we report increased GSDMB in inflammatory bowel disease, with single-cell analysis identifying epithelial specificity to inflamed colonocytes/crypt top colonocytes. Surprisingly, mechanistic experiments and transcriptome profiling reveal lack of inherent GSDMB-dependent pyroptosis in activated epithelial cells and organoids but instead point to increased proliferation and migration during in vitro wound closure, which arrests in GSDMB-deficient cells that display hyper-adhesiveness and enhanced formation of vinculin-based focal adhesions dependent on PDGF-A-mediated FAK phosphorylation. Importantly, carriage of disease-associated GSDMB SNPs confers functional defects, disrupting epithelial restitution/repair, which, altogether, establishes GSDMB as a critical factor for restoration of epithelial barrier function and the resolution of inflammation.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis , Base Sequence , Case-Control Studies , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial Cells/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HEK293 Cells , HT29 Cells , Humans , Inflammatory Bowel Diseases/genetics , Methotrexate/pharmacology , Mutation/genetics , Phosphorylation/drug effects , Polymorphism, Single Nucleotide/genetics , Pyroptosis/drug effects , Pyroptosis/genetics , Reproducibility of Results , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects , Wound Healing/drug effects , Wound Healing/genetics
5.
Cancers (Basel) ; 13(4)2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33670106

ABSTRACT

Colorectal cancer (CRC) remains the third most common cause of cancer and the second most common cause of cancer deaths worldwide. Clinicians are largely faced with advanced and metastatic disease for which few interventions are available. One poorly understood aspect of CRC involves altered organization of the actin cytoskeleton, especially at the metastatic stage of the disease. Myosin motors are crucial regulators of actin cytoskeletal architecture and remodeling. They act as mechanosensors of the tumor environments and control key cellular processes linked to oncogenesis, including cell division, extracellular matrix adhesion and tissue invasion. Different myosins play either oncogenic or tumor suppressor roles in breast, lung and prostate cancer; however, little is known about their functions in CRC. This review focuses on the functional roles of myosins in colon cancer development. We discuss the most studied class of myosins, class II (conventional) myosins, as well as several classes (I, V, VI, X and XVIII) of unconventional myosins that have been linked to CRC development. Altered expression and mutations of these motors in clinical tumor samples and their roles in CRC growth and metastasis are described. We also evaluate the potential of using small molecular modulators of myosin activity to develop novel anticancer therapies.

6.
Nat Commun ; 12(1): 262, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431859

ABSTRACT

The pathogenesis of ulcerative colitis (UC), a major type of inflammatory bowel disease, remains unknown. No model exists that adequately recapitulates the complexity of clinical UC. Here, we take advantage of induced pluripotent stem cells (iPSCs) to develop an induced human UC-derived organoid (iHUCO) model and compared it with the induced human normal organoid model (iHNO). Notably, iHUCOs recapitulated histological and functional features of primary colitic tissues, including the absence of acidic mucus secretion and aberrant adherens junctions in the epithelial barrier both in vitro and in vivo. We demonstrate that the CXCL8/CXCR1 axis was overexpressed in iHUCO but not in iHNO. As proof-of-principle, we show that inhibition of CXCL8 receptor by the small-molecule non-competitive inhibitor repertaxin attenuated the progression of UC phenotypes in vitro and in vivo. This patient-derived organoid model, containing both epithelial and stromal compartments, will generate new insights into the underlying pathogenesis of UC while offering opportunities to tailor interventions to the individual patient.


Subject(s)
Colitis, Ulcerative/pathology , Organoids/pathology , Adherens Junctions/metabolism , Cadherins/metabolism , Disease Progression , Epithelium/pathology , Fibroblasts/pathology , Humans , Inflammation/pathology , Omentum/transplantation , Phenotype , Principal Component Analysis , Sequence Analysis, RNA , Sulfonamides/pharmacology , Transcriptome/genetics , beta Catenin/metabolism
7.
iScience ; 23(11): 101719, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33205026

ABSTRACT

Colon cancer remains the third most common cause of cancer in the US, and the third most common cause of cancer death. Worldwide, colon cancer is the second most common cause of cancer and cancer deaths. At least 25% of patients still present with metastatic disease, and at least 25-30% will develop metastatic colon cancer in the course of their disease. While chemotherapy and surgery remain the mainstay of treatment, understanding the fundamental cellular niche and mechanical properties that result in metastases would facilitate both prevention and cure. Advances in biomaterials, novel 3D primary human cells, modelling using microfluidics and the ability to alter the physical environment, now offers a unique opportunity to develop and test impactful treatment.

8.
Dis Colon Rectum ; 63(11): 1559-1569, 2020 11.
Article in English | MEDLINE | ID: mdl-32868555

ABSTRACT

BACKGROUND: Colorectal cancer and IBD account for a large portion of the practice of colorectal surgery. Historical research models have provided insights into the underlying causes of these diseases but come with many limitations. OBJECTIVE: The aim of this study was to systematically review the literature regarding the advantage of organoid models in modeling benign and malignant colorectal pathology. DATA SOURCES: Sources included PubMed, Ovid-Medline, and Ovid Embase STUDY SELECTION:: Two reviewers completed a systematic review of the literature between January 2006 and January of 2020 for studies related to colon and intestinal organoids. Reviews, commentaries, protocols, and studies not performed in humans or mice were excluded. RESULTS: A total of 73 articles were included. Organoid models of colorectal disease have been rising in popularity to further elucidate the genetic, transcriptomic, and treatment response of these diseases at the individual level. Increasingly complex models utilizing coculture techniques are being rapidly developed that allow in vitro recapitulation of the disease microenvironment. LIMITATIONS: This review is only qualitative, and the lack of well utilized nomenclature in the organoid community may have resulted in the exclusion of articles. CONCLUSIONS: Historical disease models including cell lines, patient-derived tumor xenografts, and animal models have created a strong foundation for our understanding of colorectal pathology. Recent advances in 3-dimensional cell cultures, in the form of patient-derived epithelial organoids and induced human intestinal organoids have opened a new avenue for high-resolution analysis of pathology at the level of an individual patient. Recent research has shown the potential of organoids as a tool for personalized medicine with their ability to retain patient characteristics, including treatment response.


Subject(s)
Colorectal Neoplasms , Inflammatory Bowel Diseases , Models, Animal , Organoids , Xenograft Model Antitumor Assays , Animals , Cell Line , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Humans , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Mice , Precision Medicine/methods , Procedures and Techniques Utilization
9.
Integr Biol (Camb) ; 12(1): 1-11, 2020 02 22.
Article in English | MEDLINE | ID: mdl-31965190

ABSTRACT

Tumor-initiating cells (TICs), a subpopulation of cancerous cells with high tumorigenic potential and stem-cell-like properties, drive tumor progression and are resistant to conventional therapies. Identification and isolation of TICs are limited by their low frequency and lack of robust markers. Here, we characterize the heterogeneous adhesive properties of a panel of human and murine cancer cells and demonstrate differences in adhesion strength among cells, which exhibit TIC properties and those that do not. These differences in adhesion strength were exploited to rapidly (~10 min) and efficiently isolate cancerous cells with increased tumorigenic potential in a label-free manner by use of a microfluidic technology. Isolated murine and human cancer cells gave rise to larger tumors with increased growth rate and higher frequency in both immunocompetent and immunocompromised mice, respectively. This rapid and label-free TIC isolation technology has the potential to be a valuable tool for facilitating research into TIC biology and the development of more efficient diagnostics and cancer therapies.


Subject(s)
Carcinogenesis/pathology , Cell Adhesion , Cell Separation/methods , Hydrodynamics , Neoplasms/physiopathology , Neoplastic Stem Cells/pathology , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Green Fluorescent Proteins/metabolism , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, SCID , Microfluidics , Signal Transduction , Stress, Mechanical
10.
Neoplasia ; 21(3): 269-281, 2019 03.
Article in English | MEDLINE | ID: mdl-30738331

ABSTRACT

Dysfunctional inflammatory pathways are associated with an increased risk of cancer, including colorectal cancer. We have previously identified and enriched for a self-renewing, colon cancer stem cell (CCSC) subpopulation in primary sporadic colorectal cancers (CRC) and a related subpopulation in ulcerative colitis (UC) patients defined by the stem cell marker, aldehyde dehydrogenase (ALDH). Subsequent work demonstrated that CCSC-initiated tumors are dependent on the inflammatory chemokine, CXCL8, a known inducer of tumor proliferation, angiogenesis and invasion. Here, we use RNA interference to target CXCL8 and its receptor, CXCR1, to establish the existence of a functional signaling pathway promoting tumor growth initiated by sporadic and colitis CCSCs. Knocking down either CXCL8 or CXCR1 had a dramatic effect on inhibiting both in vitro proliferation and angiogenesis. Likewise, tumorigenicity was significantly inhibited due to reduced levels of proliferation and angiogenesis. Decreased expression of cycle cell regulators cyclins D1 and B1 along with increased p21 levels suggested that the reduction in tumor growth is due to dysregulation of cell cycle progression. Therapeutically targeting the CXCL8-CXCR1 signaling pathway has the potential to block sustained tumorigenesis by inhibiting both CCSC- and pCCSC-induced proliferation and angiogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Inflammation/metabolism , Interleukin-8/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Interleukin-8A/metabolism , Signal Transduction , Animals , Biomarkers , Cell Line, Tumor , Cell Proliferation , Colitis/complications , Colitis/genetics , Colitis/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Gene Dosage , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Immunophenotyping , Inflammation/complications , Inflammation/genetics , Interleukin-8/genetics , Mice , Models, Biological , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Receptors, Interleukin-8A/genetics
11.
Oncotarget ; 9(47): 28717-28730, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29983891

ABSTRACT

Ulcerative colitis (UC) is a prevalent form of inflammatory bowel disease (IBD) whose pathogenic mechanisms remain unclear. Elucidating these mechanisms is important to reduce UC symptoms and to prevent UC progression into colitis-associated colon cancer (CAC). Our goal was to develop and validate faithful, human-derived, UC models and analyze them at histologic, transcriptomic and epigenetic levels to allow mechanistic studies of UC and CAC pathogenesis. We generated patient-derived primary-organoid cultures from UC and non-IBD colonic epithelium. We phenotyped them histologically and used next-generation-sequencing approaches to profile whole transcriptomes and epigenomes of organoids and primary tissues. Tissue organization and expression of mucin 2 (MUC2) and lysozyme (LYZ) demonstrated histologic faithfulness of organoids to healthy and diseased colonic epithelium. Transcriptomic analyses showed increased expression of inflammatory pathways in UC patient-derived organoids and tissues. Profiling for active enhancers using the H3K27ac histone modification revealed UC-derived organoid enrichment for pathways indicative of gastrointestinal cancer, including S100 calcium-binding protein P (S100P), and revealed novel markers for GI cancer, including both LYZ and neuropeptide S receptor 1 (NPSR1). Immunolocalization showed increased levels of LYZ, S100P, and NPSR1 proteins in UC and CAC. In conclusion, primary colonic organoid cultures from UC and non-IBD patients can be established that faithfully represent diseased or normal colonic states. These models reveal precancerous molecular pathways that are already activated in UC. The findings demonstrate the suitability of primary organoids for dissecting UC and CAC pathogenic mechanisms and suggest new targets for therapeutic intervention.

12.
Oncotarget ; 9(16): 13048-13059, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29560130

ABSTRACT

Inflammatory bowel disease (IBD) affects one million people in the US. Ulcerative colitis (UC) is a subtype of IBD that can lead to colitis-associated cancer (CAC). In UC, the rate of CAC is 3-5-fold greater than the rate of sporadic colorectal cancer (CRC). The pathogenesis of UC and CAC are due to aberrant interactions between host immune system and microenvironment, but precise mechanisms are still unknown. In colitis and CAC, microenvironmental fibroblasts exhibit an activated, inflammatory phenotype that contributes to tumorigenesis accompanied by excessive secretion of the chemokine CXCL8. However, mechanisms regulating CXCL8 secretion are unclear. Since it is known that miRNAs regulate chemokines such as CXCL8, we queried a microRNA library for mimics affecting CXCL8 secretion. Among the identified microRNAs, miR-20a/b was further investigated as its stromal expression levels inversely correlated with the amounts of CXCL8 secreted and predicted fibroblast tumor-promoting activity. Indeed, miR-20a directly bound to the 3'UTR of CXCL8 mRNA and regulated its expression by translational repression. In vivo co-inoculation studies with CRC stem cells demonstrated that fibroblasts characterized by high miR-20a expression had reduced tumor-promoting activities. These studies reveal that in stromal fibroblasts, miR-20a modulates CXCL8 function, therefore influencing tumor latency.

13.
Oncotarget ; 8(31): 50476-50488, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881576

ABSTRACT

In sporadic colon cancer, colon cancer stem cells (CCSCs) initiate tumorigenesis and may contribute to late disease recurrences and metastases. We previously showed that aldehyde dehydrogenase (ALDH) activity (as indicated by the ALDEFLUOR® assay) is an effective marker for highly enriching CCSCs for further evaluation. Here, we used comparative transcriptome and proteome approaches to identify signaling pathways overrepresented in the CCSC population. We found overexpression of several components of the phosphoinositide 3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) signaling pathway, including PI3KR2, a regulatory subunit of PI3K. LY294002, a PI3K inhibitor, defined the contribution of the PI3K/Akt/mTOR signaling pathway in CCSCs. LY294002-treated CCSCs showed decreases in proliferation, sphere formation and self-renewal, in phosphorylation-dependent activation of Akt, and in expression of cyclin D1. Inhibition of PI3K in vivo reduced tumorigenicity, increased detection of cleaved caspase 3, an indicator of apoptosis, and elevated expression of the inflammatory chemokine, CXCL8. Collectively, these results indicate that PI3K/Akt/mTOR signaling controls CCSC proliferation and CCSC survival, and suggests that it would be useful to develop therapeutic agents that target this signaling pathway.

14.
Cell Stem Cell ; 18(2): 168-9, 2016 02 04.
Article in English | MEDLINE | ID: mdl-26849302

ABSTRACT

Administering adjuvant chemotherapy to stage II colon cancer patients is controversial due to limited benefit observed for this subpopulation. Recently, Dalerba et al. (2016) identified a subgroup of stage II patients that might benefit from adjuvant chemotherapy based on lack of CDX2 expression in their cancer stem cells.


Subject(s)
Cell Lineage , Colonic Neoplasms/pathology , CDX2 Transcription Factor , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Homeodomain Proteins/metabolism , Humans , Neoplastic Stem Cells/pathology , Trans-Activators/metabolism
15.
Proc Natl Acad Sci U S A ; 112(28): 8732-7, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26124098

ABSTRACT

A new paradigm in oncology establishes a spectrum of tumorigenic potential across the heterogeneous phenotypes within a tumor. The cancer stem cell hypothesis postulates that a minute fraction of cells within a tumor, termed cancer stem cells (CSCs), have a tumor-initiating capacity that propels tumor growth. An application of this discovery is to target this critical cell population using chemotherapy; however, the process of isolating these cells is arduous, and the rarity of CSCs makes it difficult to test potential drug candidates in a robust fashion, particularly for individual patients. To address the challenge of screening drug libraries on patient-derived populations of rare cells, such as CSCs, we have developed a drug-eluting microarray, a miniaturized platform onto which a minimal quantity of cells can adhere and be exposed to unique treatment conditions. Hundreds of drug-loaded polymer islands acting as drug depots colocalized with adherent cells are surrounded by a nonfouling background, creating isolated culture environments on a solid substrate. Significant results can be obtained by testing <6% of the cells required for a typical 96-well plate. Reliability was demonstrated by an average coefficient of variation of 14% between all of the microarrays and 13% between identical conditions within a single microarray. Using the drug-eluting array, colorectal CSCs isolated from two patients exhibited unique responses to drug combinations when cultured on the drug-eluting microarray, highlighting the potential as a prognostic tool to identify personalized chemotherapeutic regimens targeting CSCs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Drug Delivery Systems , Neoplastic Stem Cells/drug effects , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/pathology , Humans , Middle Aged , Tumor Cells, Cultured
16.
J Gastrointest Surg ; 18(5): 1040-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24643495

ABSTRACT

BACKGROUND: Colorectal cancer remains the most common gastrointestinal cancer. While screening combined with effective surgical treatment has reduced its mortality, we still do not have effective means to prevent recurrence nor to treat metastatic disease. What we know about cancer biology has gone through revolutionary changes in recent decades. The advent of the cancer stem cell theory has accelerated our understanding of the cancer cell. However, there is increasing evidence that cancer cells are influenced by their surrounding microenvironment. PURPOSE: This review divides the tumor microenvironment into four functional components-the stem cell niche, cancer stroma, immune cells, and vascular endothelia-and examines their individual and collective influence on the growth and metastasis of the colon cancer stem cell. The discussion will highlight the need to fully exploit the tumor microenvironment when designing future prognostic tools and therapies.


Subject(s)
Colonic Neoplasms/pathology , Fibroblasts/physiology , Neoplastic Stem Cells/pathology , Tumor Microenvironment , Cell Transformation, Neoplastic , Colonic Neoplasms/therapy , Dendritic Cells/physiology , Endothelial Cells/physiology , Humans , Leukocytes/physiology , Myeloid Progenitor Cells/physiology , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Paneth Cells/physiology
17.
Clin Med Res ; 10(4): 224-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22997356

ABSTRACT

OBJECTIVE: To evaluate the efficacy of transanal excision (TAE) combined with radiotherapy for rectal adenocarcinoma, assess the ability of pretreatment endoscopic ultrasound (EUS) to predict failures, and determine the prognostic value of downstaging and complete pathological response. DESIGN: Retrospective outcomes study. SETTING: Radiation oncology clinic. PARTICIPANTS: Thirty-eight patients with rectal adenocarcinoma. METHODS: The medical records of patients treated with radiotherapy from 1998 to 2008 and followed for a median of 5.9 years were reviewed. RESULTS: Kaplan-Meier estimates of freedom from selected endpoints at 5 years after treatment were: overall survival, 79%; cause-specific survival, 91%; local control, 90%; and freedom from distant metastasis, 76%. Seven patients (21%) had eventual abdominoperineal resection or lower anterior resection, four patients had local recurrence, and three patients had incomplete treatment or poor margins. T3 lesions clinically staged by EUS were a predictor of local failure (P=0.0110), but not distant metastasis (P=0.35). Patients with either a pathological or clinical T3 lesion did not have a significantly greater rate of metastasis (P=0.096). Patients who were downstaged did not have a significantly different rate of local recurrence or metastasis. Patients who experienced a complete pathological response did not have a significantly different rate of local control or distant metastasis. CONCLUSION: Patients with early-stage rectal lesions who undergo preoperative or postoperative radiation and TAE have similar outcomes to those who undergo abdominoperineal resection; local recurrence was higher for patients with T3 lesions when both were compared. Abdominal surgery should be considered for these patients. TAE is reasonable when patients are unwilling or unable to tolerate the morbidity of traditional transabdominal surgery.


Subject(s)
Adenocarcinoma/radiotherapy , Adenocarcinoma/surgery , Rectal Neoplasms/radiotherapy , Rectal Neoplasms/surgery , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Anal Canal , Endosonography , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Neoplasm, Residual , Predictive Value of Tests , Radiotherapy, Adjuvant , Rectal Neoplasms/diagnostic imaging , Rectal Neoplasms/pathology , Retrospective Studies , Survival Rate
18.
Cancer Res ; 72(19): 5091-100, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22902411

ABSTRACT

Ulcerative colitis (UC) increases the risk of colorectal cancer (CRC), but the mechanisms involved in colitis-to-cancer transition (CCT) are not well understood. CCT may involve a inflammation-dysplasia-carcinoma progression sequence compared with the better characterized adenoma-carcinoma progression sequence associated with sporadic CRC. One common thread may be activating mutations in components of the Wnt/ß-catenin signaling pathway, which occur commonly as early events in sporadic CRC. To examine this hypothesis, we evaluated possible associations between Wnt/ß-catenin signaling and CCT based on the cancer stem cell (CSC) model. Wnt/ß-catenin immunostaining indicated that UC patients have a level of Wnt-pathway-active cells that is intermediate between normal colon and CRC. These UC cells exhibiting activation of the Wnt pathway constituted a major subpopulation (52% + 7.21) of the colonic epithelial cells positive for aldehyde dehydrogenase (ALDH), a putative marker of precursor colon CSC (pCCSC). We further fractionated this subpopulation of pCCSC using a Wnt pathway reporter assay. Over successive passages, pCCSCs with the highest Wnt activity exhibited higher clonogenic and tumorigenic potential than pCCSCs with the lowest Wnt activity, thereby establishing the key role of Wnt activity in driving CSC-like properties in these cells. Notably, 5/20 single cell injections of high-Wnt pCCSC resulted in tumor formation, suggesting a correlation with CCT. Attenuation of Wnt/ß-catenin in high-Wnt pCCSC by shRNA-mediated downregulation or pharmacological inhibition significantly reduced tumor growth rates. Overall, the results of our study indicates (i) that early activation of Wnt/ß-catenin signaling is critical for CCT and (ii) that high levels of Wnt/ß-catenin signaling can further demarcate high-ALDH tumor-initiating cells in the nondysplastic epithelium of UC patients. As such, our findings offer plausible diagnostic markers and therapeutic target in the Wnt signaling pathway for early intervention in CCT.


Subject(s)
Colitis/metabolism , Colonic Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Wnt Signaling Pathway , Aldehyde Dehydrogenase/metabolism , Animals , Blotting, Western , Cell Transformation, Neoplastic , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , RNA Interference , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transplantation, Heterologous , Tumor Burden , Tumor Cells, Cultured , beta Catenin/genetics , beta Catenin/metabolism
19.
Methods Mol Biol ; 916: 373-85, 2012.
Article in English | MEDLINE | ID: mdl-22914954

ABSTRACT

Aldehyde dehydrogenase (ALDH) can be used as a marker to isolate, propagate, and track normal and cancerous human colon stem cells. To determine their tumorigenic potential, tissues obtained from proximal (normal counterpart) and distal (cancerous) colon of colon cancer patients are implanted into NOD-SCID mice. In parallel, ALDH(high) and ALDH(low) cells are isolated via Florescence Associated Cell Sorting (FACS) after the dissociation of distal and proximal colon tissues into a single-cell suspension. Flow cytometry for ALDH(high) and ALDH(low) cells is possible with the ALDEFLUOR assay. Following cell sorting, ALDH-enriched cells are tested for their tumorigenic potential in vivo as xenografts. Owing to cancer stem cell properties, ALDH(high) cells could be propagated in vivo by serial passaging of the human tissue as xenografts and in vitro as suspension cultures called sphere cultures. In this unit, all the above-mentioned methods to isolate and propagate colon cancer stem cells using ALDH as a stem cell marker are described in detail.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Biomarkers, Tumor/metabolism , Colon/cytology , Colon/pathology , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Stem Cells/enzymology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Colon/enzymology , Flow Cytometry , Humans , Immunohistochemistry , Matrix Metalloproteinase 13/metabolism , Mice , Stem Cells/cytology , Stem Cells/pathology , Tissue Fixation
20.
Oncol Rev ; 5(3): 177-184, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21949574

ABSTRACT

Antiangiogenic therapy has shown promise in the treatment of patients with hepatocellular carcinoma (HCC). Bevacizumab, sorafenib, and sunitinib showed efficacy in patients with HCC; and sorafenib is approved by the FDA for treatment of this cancer. In practice, the clinical benefit of these agents has been heterogeneous; and in patients who do respond, the benefit is modest and/or short-lived. Recent advances in the molecular understanding of tumor angiogenesis along with the rapid development of targeted drug discovery have made it possible to explore novel combination therapy for HCC. We review the clinical trial results, discuss possible molecular mechanisms of resistance, and suggest novel combinations with antiangiogenic therapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...