Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
J Natl Cancer Cent ; 2(4): 235-242, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36568283

ABSTRACT

"Screening" is a search for preclinical, asymptomatic disease, including cancer. Widespread cancer screening has led to large increases in early-stage cancers and pre-cancers. Ubiquitous public messages emphasize the potential benefits to screening for these lesions based on the underlying assumption that treating cancer at early stages before spread to other organs should make it easier to treat and cure, using more tolerable interventions. The intuition is so strong that public campaigns are sometimes launched without conducting definitive trials directly comparing screening to usual care. An effective cancer screening test should not only increase the incidence of early-stage preclinical disease but should also decrease the incidence of advanced and metastatic cancer, as well as a subsequent decrease in cancer-related mortality. Otherwise, screening efforts may be uncovering a reservoir of non-progressive and very slowly progressive lesions that were not destined to cause symptoms or suffering during the person's remaining natural lifespan: a phenomenon known as "overdiagnosis." We provide here a qualitative review of cancer overdiagnosis and discuss specific examples due to extensive population-based screening, including neuroblastoma, prostate cancer, thyroid cancer, lung cancer, melanoma, and breast cancer. The harms of unnecessary diagnosis and cancer therapy call for a balanced presentation to people considering undergoing screening, even with a test of accepted benefit, with a goal of informed decision-making. We also discuss proposed strategies to mitigate the adverse sequelae of overdiagnosis.

2.
Front Oncol ; 10: 1201, 2020.
Article in English | MEDLINE | ID: mdl-32850349
3.
Front Oncol ; 10: 186, 2020.
Article in English | MEDLINE | ID: mdl-32231993

ABSTRACT

The term "big data" refers broadly to large volumes of data, often gathered from several sources, that are then analyzed, for example, for predictive analytics. Combining and mining genetic data from varied sources including clinical genetic testing, for example, electronic health records, what might be termed as "recreational" genetic testing such as ancestry testing, as well as research studies, provide one type of "big data." Challenges and cautions in analyzing big data include recognizing the lack of systematic collection of the source data, the variety of assay technologies used, the potential variation in classification and interpretation of genetic variants. While advanced technologies such as microarrays and, more recently, next-generation sequencing, that enable testing an individual's DNA for thousands of genes and variants simultaneously are briefly discussed, attention is focused more closely on challenges to analysis of the massive data generated by these genomic technologies. The main theme of this review is to evaluate challenges associated with big data in general and specifically to bring the sophisticated technology of genetic/genomic testing down to the individual level, keeping in mind the human aspect of the data source and considering where the impact of the data will be translated and applied. Considerations in this "humanizing" process include providing adequate counseling and consent for genetic testing in all settings, as well as understanding the strengths and limitations of assays and their interpretation.

4.
Am J Epidemiol ; 189(1): 27-33, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31595954

ABSTRACT

Advances in cancer screening methods have opened avenues for incidental findings and cancer overdiagnosis. We performed a secondary analysis of the National Lung Screening Trial (enrollment from 2002-2004), a randomized controlled trial comparing low-dose computed tomography (LDCT; n = 26,722) with chest radiography (CXR; n = 26,732) for lung cancer detection, to examine incidental findings related to thyroid cancer (ThCa). Three screening rounds were included, and median follow-up was 6.6 years for LDCT and 6.5 years for CXR. Radiologists reported lung and non-lung-related abnormalities. In the LDCT arm, 5.7%, 4.7%, and 4.5% of participants had abnormalities above the diaphragm (AADs) detected at baseline, year 1, and year 2, respectively, compared with 2.3%, 1.5%, and 1.3% in the CXR arm. In the LDCT arm, 205 AADs (7.0%) were thyroid-related. Overall, 60 ThCas were reported, 35 in the LDCT arm and 25 in the CXR arm (P = 0.2). In the LDCT arm, participants with a prior AAD had a 7.8-fold increased risk (95% confidence interval: 4.0, 15.1) of ThCa compared with those who did not have an AAD. Early and persistent excess of ThCas diagnosed earlier in the LDCT arm suggests overdiagnosis. The use of sensitive screening modalities for early detection of lung cancer might result in the discovery of thyroid incidentalomas.


Subject(s)
Early Detection of Cancer/statistics & numerical data , Lung Neoplasms/diagnosis , Radiography, Thoracic/statistics & numerical data , Thyroid Neoplasms/diagnosis , Tomography, X-Ray Computed/statistics & numerical data , Aged , Female , Humans , Incidental Findings , Lung Neoplasms/etiology , Male , Medical Overuse , Middle Aged , Prospective Studies , Smoking/adverse effects , Thyroid Neoplasms/epidemiology , United States/epidemiology
5.
Front Oncol ; 9: 765, 2019.
Article in English | MEDLINE | ID: mdl-31555578

ABSTRACT

Obesity is associated with increased risk of breast and other cancers. However, the complexity of the underlying mechanisms, together with the interplay of diet and physical activity-contributing to energy balance-and the role of adipose tissue, pose challenges to our understanding of the basis of this increased risk. Epidemiologic studies have documented a higher obesity prevalence in US black women compared to white women. Elucidation of the contribution of potential biological differences among racially distinct groups to their differences in breast cancer (BC) risk and mortality have been topics of considerable interest in recent years. The racial and ethnic variation in body fat distribution may account for at least part of the differences in breast cancer rates in these populations. Yet, while black women exhibit higher rates of obesity compared to white women, this does not translate directly into higher rates of BC. In fact, overall, BC in black women occurs with a lower incidence than BC in white women. Obesity is a known risk factor for postmenopausal breast cancer, and growing evidence suggests that abdominal obesity, also known as central obesity, may increase risk for triple negative breast cancer, which is more common in premenopausal women. The positive association of postmenopausal BC risk and specifically estrogen receptor (ER)-positive BC, is presumably due largely to accumulation of estrogen in the adipose tissue of the breast and other tissues. Of the two main types of adipose tissue-subcutaneous and visceral-visceral adipocytes are more active metabolically. Such adipose tissue harbors multiple molecular entities that promote carcinogenesis: endocrine molecules/hormones, immunologic factors, inflammatory cytokines, metabolic alterations, and other components of the microenvironment. Expression of these culpable entities is largely regulated by epigenetic mechanisms. The interrelationship between these entities and drivers of epigenetic alteration are critical to the regulation of pathways connecting obesity and cancer risk. Initiatives to counteract the carcinogenic effects of obesity have primarily involved modulation of energy balance by diet. However, targeting of specific molecular abnormalities characterizing adiposity offers an alternative approach to preventing cancer. Our goal in this review is to first discuss the major mechanisms contributing to the obesity-breast cancer link. We will also consider race, specifically black/white differences, as they relate to the association of obesity with breast cancer risk. Then we will enumerate strategies targeting these mechanisms to reduce BC risk, in large part by way of dietary interventions with potential to mitigate the cancer-promoting components of adiposity.

6.
Cancer Res ; 79(20): 5140-5145, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31337654

ABSTRACT

The recent explosion and ease of access to large-scale genomics data is intriguing. However, serious obstacles exist to the optimal management of the entire spectrum from data production in the laboratory through bioinformatic analysis to statistical evaluation and ultimately clinical interpretation. Beyond the multitude of technical issues, what stands out the most is the absence of adequate communication among the specialists in these domains. Successful interdisciplinary collaborations along the genomics pipeline extending from laboratory experiments to bioinformatic analyses to clinical application are notable in large scale, well managed projects such as The Cancer Genome Atlas. However, in certain settings in which the various experts perform their specialized research activities in isolation, the siloed approach to their research contributes to the generation of questionable genomic interpretations. Such situations are particularly concerning when the ultimate endpoint involves genetic/genomic interpretations that are intended for clinical applications. In spite of the fact that clinicians express interest in gaining a better understanding of clinical genomic applications, the lack of communication from upstream experts leaves them with a serious level of discomfort in applying such genomic knowledge to patient care. This discomfort is especially evident among healthcare providers who are not trained as geneticists, in particular primary care physicians. We offer some initiatives that have potential to address this problem, with emphasis on improved and ongoing communication among all the experts in these fields, constituting a comprehensive genomic "pipeline" from laboratory to patient.


Subject(s)
Genomics , High-Throughput Nucleotide Sequencing , Interdisciplinary Communication , Advisory Committees , Artifacts , Communication Barriers , Comprehension , Data Analysis , Electronic Health Records , Genetic Counseling , Humans , Primary Health Care , Professional Competence , Reproducibility of Results , Translational Research, Biomedical
7.
Cancer Prev Res (Phila) ; 11(4): 203-214, 2018 04.
Article in English | MEDLINE | ID: mdl-29453232

ABSTRACT

Obesity, a cause of subclinical inflammation, is a risk factor for the development of postmenopausal breast cancer and is associated with poorer cancer outcomes. Docosahexaenoic acid (DHA), an omega-3 fatty acid, possesses anti-inflammatory properties. We hypothesized that treatment with DHA would reduce the expression of proinflammatory genes and aromatase, the rate-limiting enzyme for estrogen biosynthesis, in benign breast tissue of overweight/obese women. A randomized, placebo-controlled, double-blind phase II study of DHA given for 12 weeks to overweight/obese women with a history of stage I-III breast cancer, DCIS/LCIS, Paget's disease, or proliferative benign breast disease was carried out. In this placebo controlled trial, the primary objective was to determine whether DHA (1,000 mg by mouth twice daily) reduced breast tissue levels of TNFα. Secondary objectives included evaluation of the effect of DHA on breast tissue levels of COX-2, IL1ß, aromatase, white adipose tissue inflammation, and gene expression by RNA-seq. Red blood cell fatty acid levels were measured to assess compliance. From July 2013 to November 2015, 64 participants were randomized and treated on trial (32 women per arm). Increased levels of omega-3 fatty acids in red blood cells were detected following treatment with DHA (P < 0.001) but not placebo. Treatment with DHA did not alter levels of TNFα (P = 0.71), or other biomarkers including the transcriptome in breast samples. Treatment with DHA was overall well-tolerated. Although compliance was confirmed, we did not observe changes in the levels of prespecified biomarkers in the breast after treatment with DHA when compared with placebo. Cancer Prev Res; 11(4); 203-14. ©2018 AACRSee related editorial by Fabian and Kimler, p. 187.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Docosahexaenoic Acids/therapeutic use , Fibrocystic Breast Disease/drug therapy , Precancerous Conditions/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Double-Blind Method , Female , Fibrocystic Breast Disease/genetics , Fibrocystic Breast Disease/pathology , Follow-Up Studies , Gene Expression Profiling , Humans , Middle Aged , Neoplasm Invasiveness , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Prognosis
8.
Cancer Treat Rev ; 55: 1-9, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28262606

ABSTRACT

Most patients with ductal carcinoma in situ of the breast (DCIS) are eligible for breast conservation treatment. The key management decision is whether to add radiotherapy and/or endocrine therapy to minimize the risk of a subsequent recurrence. Recent analyses indicating a lack of benefit in terms of breast cancer-associated mortality have suggested that more conservative approaches, omitting adjuvant therapy or even surgery, may be advisable in selected patients. These mortality observations are directly influenced by widespread use of mammographic screening which has opened a Pandora's box of subclinical DCIS and early invasive lesions. Confusion as to how aggressively such possibly indolent lesions should be treated has led to misunderstandings among patients and medical professionals. While awaiting further prospective evidence from clinical trials, we endorse an active treatment of DCIS as the standard of care. Our rationale is twofold: invasive recurrences are associated with an increase in breast cancer mortality, which is not the only relevant endpoint for DCIS. The benefit of complete surgical excision, adjuvant radiotherapy and endocrine treatment in preventing recurrence and invasive progression has been demonstrated in DCIS. The challenge now is how to identify DCIS patients who will not progress to invasive carcinoma even without complete excision and, at the other extreme, those patients at the highest risk who require mastectomy for local control. The current controversies over whether and which adjuvant therapy should be implemented can at least in part be addressed by developing effective doctor-patient communications that enable mutual understanding about the management of this biologically heterogeneous disease.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/therapy , Chemotherapy, Adjuvant , Radiotherapy, Adjuvant , Female , Humans , Mastectomy, Segmental
9.
Expert Rev Clin Pharmacol ; 9(10): 1263-1272, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27583816

ABSTRACT

INTRODUCTION: Raloxifene is an estrogen receptor modulator which competes with estrogens for binding to the estrogen receptor. Based on the results of the STAR (Study of Tamoxifen And Raloxifene) trial, raloxifene has been approved by the U.S. Food and Drug Administration for the reduction of breast cancer (BC) risk in postmenopausal women at increased risk. Areas covered: This analysis reviews the activity of raloxifene and the clinical trials for non-BC indications which led to investigate its use as BC preventive agent. We review the trial establishing its efficacy for BC prevention and the meta-analyses including different SERMs for BC prevention. Expert commentary: Compared with tamoxifen, raloxifene has shown a slightly lower efficacy in reducing BC risk and a better safety profile. Raloxifene also offers to postmenopausal women a benefit in terms of osteoporosis. Future research should investigate its use in premenopausal women and in association with other preventive agents.

11.
Semin Oncol ; 43(1): 36-48, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26970123

ABSTRACT

Cancer is a complex category of diseases caused in large part by genetic or genomic, transcriptomic, and epigenetic or epigenomic alterations in affected cells and the surrounding microenvironment. Carcinogenesis reflects the clonal expansion of cells that progressively acquire these genetic and epigenetic alterations-changes that, in turn, lead to modifications at the RNA level. Gradually advancing technology and most recently, the advent of next-generation sequencing (NGS), combined with bioinformatics analytic tools, have revolutionized our ability to interrogate cancer cells. The ultimate goal is to apply these high-throughput technologies to the various aspects of clinical cancer care: cancer-risk assessment, diagnosis, as well as target identification for treatment and prevention. In this article, we emphasize how the knowledge gained through large-scale omics-oriented approaches, with a focus on variations at the level of nucleic acids, can inform the field of chemoprevention.


Subject(s)
Genetic Variation , Genomics/methods , Neoplasms/genetics , Neoplasms/prevention & control , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Epigenesis, Genetic , Gene Dosage , Gene Expression Profiling/methods , Humans , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA , Sequence Analysis, RNA , Transcriptome
12.
Semin Oncol ; 43(1): 123-133, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26970131

ABSTRACT

Multiple epidemiologic studies have documented an association between the anti-diabetic agent metformin and reduced cancer incidence and mortality. However, this effect has not been consistently demonstrated in animal models or more recent epidemiological studies. The purpose of this paper is to examine metformin's chemopreventive potential by reviewing relevant mechanisms of action, preclinical evidence of efficacy, updated epidemiologic evidence after correction for potential biases and confounders, and recently completed and ongoing clinical trials. Although repurposing drugs with well described mechanisms of action and safety profiles is an appealing strategy for cancer prevention, there is no substitute for well executed late phase clinical trials to define efficacy and populations that are most likely to benefit from an intervention.


Subject(s)
Anticarcinogenic Agents/pharmacology , Drug Repositioning , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/therapeutic use , Chemoprevention , Diabetes Mellitus/drug therapy , Humans , Hypoglycemic Agents/therapeutic use , Incidence , Metformin/therapeutic use , Neoplasms/epidemiology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
13.
Semin Oncol ; 43(1): 161-172, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26970135

ABSTRACT

The potential of the immune system to recognize and reject tumors has been investigated for more than a century. However, only recently impressive breakthroughs in cancer immunotherapy have been seen with the use of checkpoint inhibitors. The experience with various immune-based strategies in the treatment of late cancer highlighted the importance of negative impact advanced disease has on immunity. Consequently, use of immune modulation for cancer prevention rather than therapy has gained considerable attention, with many promising results seen already in preclinical and early clinical studies. Although not without challenges, these results provide much excitement and optimism that successful cancer immunoprevention could be within our reach. In this review we will discuss the current state of predominantly primary and secondary cancer immunoprevention, relevant research, potential barriers, and future directions.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Breast Neoplasms/prevention & control , Colorectal Neoplasms/immunology , Colorectal Neoplasms/prevention & control , Immunotherapy , Pancreatic Neoplasms/immunology , Adaptive Immunity , Animals , Autoantigens/immunology , Colorectal Neoplasms/genetics , Female , Frameshift Mutation , Hepatitis B Vaccines/therapeutic use , Humans , Immunity, Innate , Immunologic Surveillance , Lactalbumin/immunology , Mammaglobin A/immunology , Papillomavirus Vaccines/therapeutic use , Receptor, ErbB-2/immunology , Telomerase/antagonists & inhibitors , Telomerase/immunology
14.
Trends Cancer ; 2(12): 713-722, 2016 12.
Article in English | MEDLINE | ID: mdl-28138568

ABSTRACT

In this review, we address selected areas that are central to the state-of-the-art of cancer prevention science. The emphasis on prevention as a viable and critical approach to decreasing cancer mortality has gained traction in recent years, evidenced by its inclusion in the US Vice President's Cancer Initiative (also termed 'Moonshot'). Cancer prevention occurs by arresting, slowing down, or reversing the carcinogenic process before invasion into surrounding tissue or by avoiding or blocking causative exposure. An important challenge is to identify individuals who will benefit most from preventive interventions with the least possible harm. Preventive interventions range from avoiding known carcinogens (e.g., tobacco or asbestos) to intervening with anticarcinogenic strategies (behavioral modifications , such as diet and exercise; medications; nutritional agents; and vaccination against causative agents). Here, we focus on active intervention with measures involving pharmaceutical and immunological agents.


Subject(s)
Neoplasms/prevention & control , Primary Prevention , Animals , Anticarcinogenic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Diet , Humans
15.
J Natl Cancer Inst ; 107(12): djv259, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26420882

ABSTRACT

BACKGROUND: Over 25 years, the National Cancer Institute's Division of Cancer Prevention has entered some 800 agents into a chemopreventive agent testing program. Two critical steps involve: 1) in vitro/in vivo morphologic assays and 2) animal tumor assays (incidence/multiplicity reduction). We sought to determine how accurately the earlier-stage (morphologic) assays predict efficacy in the later-stage (animal tumor) assays. METHODS: Focusing on 210 agents tested in both morphologic and animal tumor assays, we carried out statistical modeling of how well the six most commonly used morphologic assays predicted drug efficacy in animal tumor assays. Using multimodel inference, three statistical models were generated to evaluate the ability of these six morphologic assays to predict tumor outcomes in three different sets of animal tumor assays: 1) all tumor types, 2) mammary cancer only, and 3) colon cancer only. Using this statistical modeling approach, each morphologic assay was assigned a value reflecting how strongly it predicted outcomes in each of the three different sets of animal tumor assays. RESULTS: We demonstrated differences in the predictive value of specific morphologic assays for positive animal tumor assay results. Some of the morphologic assays were strongly predictive of meaningful positive efficacy outcomes in animal tumor assays representing specific cancer types, particularly the aberrant crypt focus (ACF) assay for colon cancer. Moreover, less strongly predictive assays can be combined and sequenced, resulting in enhanced composite predictive ability. CONCLUSIONS: Predictive models such as these could be used to guide selection of preventive agents as well as morphologic and animal tumor assays, thereby improving the efficiency of our approach to chemopreventive agent development.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Line, Tumor/drug effects , Models, Statistical , Neoplasms/pathology , Neoplasms/prevention & control , Primary Prevention/methods , Animals , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cell Line, Tumor/pathology , Chemoprevention/methods , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Female , Humans , Mice , Predictive Value of Tests , Rats , Treatment Outcome
16.
Expert Opin Drug Saf ; 14(10): 1573-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26359221

ABSTRACT

INTRODUCTION: Epidemiological evidence suggests an increased incidence of cancer in obese, prediabetic, and diabetic patients and a reduced risk of cancer incidence and mortality in diabetic patients on metformin compared with other antidiabetic drugs. In vitro studies support the efficacy of metformin in cancer therapy and prevention. Although metformin seems to be promising as a cancer chemopreventive or therapeutic drug, the principal consideration is whether metformin will be effective in cancer clinical trials for nondiabetic subjects or only in diabetics or subjects with insulin resistance. Safety of metformin is even more important in treating nondiabetic patients. AREAS COVERED: The present review focuses on epidemiological data and clinical trials testing the efficacy of metformin on cancer, the safety in nondiabetic patients and the future development of this promising drug. EXPERT OPINION: Meta-analyses of epidemiological in which metformin treatment has been used for diabetic patients show a positive trend for benefit; nevertheless, clinical data outcomes are preliminary and the results of ongoing trials are awaited. The different types of cancer, heterogeneity of populations and presence of comorbidity make it difficult to determine the benefits of metformin in cancer prevention and treatment.


Subject(s)
Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Neoplasms/prevention & control , Diabetes Mellitus/drug therapy , Humans , Incidence , Insulin Resistance , Neoplasms/epidemiology , Neoplasms/etiology , Obesity/complications , Prediabetic State/complications , Risk Factors
18.
J Natl Cancer Inst ; 107(3)2015 Mar.
Article in English | MEDLINE | ID: mdl-25713150

ABSTRACT

The Journal of the National Cancer Institute (JNCI), with its broad coverage of bench research, epidemiologic studies, and clinical trials, has a long history of publishing practice-changing studies in cancer prevention and public health. These include studies of tobacco cessation, chemoprevention, and nutrition. The landmark Breast Cancer Prevention Trial (BCPT)-the first large trial to prove efficacy of a preventive medication for a major malignancy-was published in the Journal, as were key ancillary papers to the BCPT. Even when JNCI was not the publication venue for the main trial outcomes, conceptual and design discussions leading to the trial as well as critical follow-up analyses based on trial data from the Prostate Cancer Prevention Trial (PCPT) and the Selenium and Vitamin E Chemoprevention Trial (SELECT) were published in the Journal. The Journal has also published important evidence on very charged topics, such as the purported link between abortion and breast cancer risk. In summary, JNCI has been at the forefront of numerous major publications related to cancer prevention.


Subject(s)
Biomedical Research/trends , National Cancer Institute (U.S.) , Neoplasms/prevention & control , Public Health , Breast Neoplasms/prevention & control , Chemoprevention , Female , Food , Humans , Male , Primary Prevention , Prostatic Neoplasms/prevention & control , Secondary Prevention , Selenium , Smoking Cessation , United States , Vitamin E
19.
Clin Cancer Res ; 20(14): 3672-82, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25028506

ABSTRACT

PURPOSE: Local transdermal therapy to the breast may achieve effective target-organ drug delivery, while diminishing systemic effects. We conducted a randomized, double-blind, placebo-controlled phase II trial comparing transdermal 4-hydroxytamoxifen gel (4-OHT) to oral tamoxifen (oral-T) in women with ductal carcinoma in situ (DCIS). METHODS: Twenty-seven pre- and postmenopausal women were randomized to 4-OHT (4 mg/day) or oral-T (20 mg/day) for 6 to 10 weeks before surgery. Plasma, nipple aspirate fluid, and breast adipose tissue concentrations of tamoxifen and its major metabolites were determined by liquid chromatography/tandem mass spectrometry. The primary endpoint was Ki67 labeling in DCIS lesions, measured by immunohistochemistry. In plasma, insulin-like growth factor-1 (IGFI), sex hormone-binding globulin (SHBG), and coagulation protein concentrations were determined. RESULTS: Posttherapy Ki67 decreased by 3.4% in the 4-OHT and 5.1% in the oral-T group (P ≤ 0.03 in both, between-group P = 0. 99). Mean plasma 4-OHT was 0.2 and 1.1 ng/mL in 4-OHT and oral groups, respectively (P = 0.0003), whereas mean breast adipose tissue concentrations of 4-OHT were 5.8 ng/g in the 4-OHT group and 5.4 ng/g in the oral group (P = 0.88). There were significant increases in plasma SHBG, factor VIII, and von Willebrand factor and a significant decrease in plasma IGFI with oral-T, but not with 4-OHT. The incidence of hot flashes was similar in both groups. CONCLUSIONS: The antiproliferative effect of 4-OHT gel applied to breast skin was similar to that of oral-T, but effects on endocrine and coagulation parameters were reduced. These findings support the further evaluation of local transdermal therapy for DCIS and breast cancer prevention.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Breast Neoplasms/drug therapy , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Tamoxifen/analogs & derivatives , Administration, Cutaneous , Administration, Oral , Aged , Antineoplastic Agents, Hormonal/pharmacokinetics , Biomarkers, Tumor/blood , Double-Blind Method , Female , Humans , Middle Aged , Tamoxifen/administration & dosage , Tamoxifen/pharmacokinetics , Treatment Outcome
20.
Cancer Prev Res (Phila) ; 7(9): 867-85, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24985407

ABSTRACT

Previous meta-analyses have shown that the antidiabetic agent metformin is associated with reduced cancer incidence and mortality. However, this effect has not been consistently demonstrated in animal models and recent epidemiologic studies. We performed a meta-analysis with a focus on confounders and biases, including body mass index (BMI), study type, and time-related biases. We identified 71 articles published between January 1, 1966, and May 31, 2013, through Pubmed, ISI Web of Science (Science Citation Index Expanded), Embase, and the Cochrane library that were related to metformin and cancer incidence or mortality. Study characteristics and outcomes were abstracted for each study that met inclusion criteria. We included estimates from 47 independent studies and 65,540 cancer cases in patients with diabetes. Overall cancer incidence was reduced by 31% [summary relative risk (SRR), 0.69; 95% confidence interval (CI), 0.52-0.90], although between-study heterogeneity was considerable (I(2) = 88%). Cancer mortality was reduced by 34% (SRR, 0.66; 95% CI, 0.54-0.81; I(2) = 21%). BMI-adjusted studies and studies without time-related biases also showed significant reduction in cancer incidence (SRR, 0.82; 95% CI, 0.70-0.96 with I(2) = 76% and SRR, 0.90; 95% CI, 0.89-0.91 with I(2) = 56%, respectively), albeit with lesser magnitude (18% and 10% reduction, respectively). However, studies of cancer mortality and individual organ sites did not consistently show significant reductions across all types of analyses. Although these associations may not be causal, our results show that metformin may reduce cancer incidence and mortality in patients with diabetes However, the reduction seems to be of modest magnitude and not affecting all populations equally. Clinical trials are needed to determine if these observations apply to nondiabetic populations and to specific organ sites.


Subject(s)
Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Neoplasms/epidemiology , Diabetes Mellitus, Type 2/drug therapy , Humans , Risk
SELECTION OF CITATIONS
SEARCH DETAIL
...