Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Ann Oncol ; 34(9): 813-825, 2023 09.
Article in English | MEDLINE | ID: mdl-37330052

ABSTRACT

BACKGROUND: The isolation of cell-free DNA (cfDNA) from the bloodstream can be used to detect and analyze somatic alterations in circulating tumor DNA (ctDNA), and multiple cfDNA-targeted sequencing panels are now commercially available for Food and Drug Administration (FDA)-approved biomarker indications to guide treatment. More recently, cfDNA fragmentation patterns have emerged as a tool to infer epigenomic and transcriptomic information. However, most of these analyses used whole-genome sequencing, which is insufficient to identify FDA-approved biomarker indications in a cost-effective manner. PATIENTS AND METHODS: We used machine learning models of fragmentation patterns at the first coding exon in standard targeted cancer gene cfDNA sequencing panels to distinguish between cancer and non-cancer patients, as well as the specific tumor type and subtype. We assessed this approach in two independent cohorts: a published cohort from GRAIL (breast, lung, and prostate cancers, non-cancer, n = 198) and an institutional cohort from the University of Wisconsin (UW; breast, lung, prostate, bladder cancers, n = 320). Each cohort was split 70%/30% into training and validation sets. RESULTS: In the UW cohort, training cross-validated accuracy was 82.1%, and accuracy in the independent validation cohort was 86.6% despite a median ctDNA fraction of only 0.06. In the GRAIL cohort, to assess how this approach performs in very low ctDNA fractions, training and independent validation were split based on ctDNA fraction. Training cross-validated accuracy was 80.6%, and accuracy in the independent validation cohort was 76.3%. In the validation cohort where the ctDNA fractions were all <0.05 and as low as 0.0003, the cancer versus non-cancer area under the curve was 0.99. CONCLUSIONS: To our knowledge, this is the first study to demonstrate that sequencing from targeted cfDNA panels can be utilized to analyze fragmentation patterns to classify cancer types, dramatically expanding the potential capabilities of existing clinically used panels at minimal additional cost.


Subject(s)
Cell-Free Nucleic Acids , Circulating Tumor DNA , Prostatic Neoplasms , Male , Humans , Circulating Tumor DNA/genetics , Mutation , Prostatic Neoplasms/genetics , Cell-Free Nucleic Acids/genetics , Gene Expression Profiling , Biomarkers, Tumor/genetics
2.
Ir J Psychol Med ; 39(1): 74-84, 2022 03.
Article in English | MEDLINE | ID: mdl-30777583

ABSTRACT

OBJECTIVES: Our principle objective was to examine the personal and professional impact of service user (SU) suicide on mental health professionals (MHPs). We also wished to explore putative demographic or clinical factors relating to SUs or MPHs that could influence the impact of SU suicide for MHPs and explore factors MHPs report as helpful in reducing distress following SU suicide. METHODS: A mixed-method questionnaire with quantitative and thematic analysis was utilised. RESULTS: Quantitative data indicated SU suicide was associated with personal and professional distress with sadness (79.5%), shock (74.5%) and surprise (68.7%) particularly evident with these phenomena lasting less than a year for more than 90% of MHPs. MHPs also reported guilt, reduced self-confidence and a fear of negative publicity. Thematic analysis indicated that some MHPs had greater expertise when addressing SU suicidal ideation and in supporting colleagues after experiencing a SU suicide. Only 17.7% of MHPs were offered formal support following SU suicide. CONCLUSION: SU suicide impacts MHPs personally and professionally in both a positive and negative fashion. A culture and clear pathway of formal support for MHPs to ascertain the most appropriate individualised support dependent on the distress they experience following SU suicide would be optimal.


Subject(s)
Mental Health , Suicide , Health Personnel/psychology , Humans , Self Concept , Suicide/psychology , Surveys and Questionnaires
3.
Neurochirurgie ; 68(1): 69-85, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33864773

ABSTRACT

IMPORTANCE: Brain metastasis (BM) is the most common malignant intracranial neoplasm in adults with over 100,000 new cases annually in the United States and outnumbering primary brain tumors 10:1. OBSERVATIONS: The incidence of BM in adult cancer patients ranges from 10-40%, and is increasing with improved surveillance, effective systemic therapy, and an aging population. The overall prognosis of cancer patients is largely dependent on the presence or absence of brain metastasis, and therefore, a timely and accurate diagnosis is crucial for improving long-term outcomes, especially in the current era of significantly improved systemic therapy for many common cancers. BM should be suspected in any cancer patient who develops new neurological deficits or behavioral abnormalities. Gadolinium enhanced MRI is the preferred imaging technique and BM must be distinguished from other pathologies. Large, symptomatic lesion(s) in patients with good functional status are best treated with surgery and stereotactic radiosurgery (SRS). Due to neurocognitive side effects and improved overall survival of cancer patients, whole brain radiotherapy (WBRT) is reserved as salvage therapy for patients with multiple lesions or as palliation. Newer approaches including multi-lesion stereotactic surgery, targeted therapy, and immunotherapy are also being investigated to improve outcomes while preserving quality of life. CONCLUSION: With the significant advancements in the systemic treatment for cancer patients, addressing BM effectively is critical for overall survival. In addition to patient's performance status, therapeutic approach should be based on the type of primary tumor and associated molecular profile as well as the size, number, and location of metastatic lesion(s).


Subject(s)
Brain Neoplasms , Radiosurgery , Adult , Aged , Brain Neoplasms/surgery , Cranial Irradiation , Humans , Prognosis , Quality of Life , Retrospective Studies , Salvage Therapy
4.
Breast Cancer Res Treat ; 113(1): 189-96, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18256926

ABSTRACT

BACKGROUND: In 2001, the National Cancer Institute's Surveillance, Epidemiology, and End Results (SEER) program established Residual Tissue Repositories (RTR) in the Hawaii, Iowa, and Los Angeles Tumor Registries to collect discarded tissue blocks from pathologic laboratories within their catchment areas. To validate the utility of the RTR for supplementing SEER's central database, we assessed human epidermal growth factor receptor-2 (HER2) and estrogen receptor expression (ER) in a demonstration project. MATERIALS: Using a prepared set of tissue microarrays (TMAs) residing in the Hawaii Tumor Registry (HTR), we performed standard immunohistochemistry. Breast cancers in the TMA were diagnosed in 1995, followed through 2006, and linked to SEER's main database. RESULTS: The TMA included 354 cases, representing 51% of 687 breast cancers in the HTR (1995). The HTR and TMA cases were similar with respect to patient demographics and tumor characteristics. Seventy-six percent (76%, 268 of 354) of TMA cases were HER2+ and/or ER+, i.e., 28 HER2+ER-, 12 HER2+ER+, and 228 HER2-ER+. There were 67 HER2-ER- cases and 19 were unclassified. Age distributions at diagnosis were bimodal with dominant early-onset modes for HER2+ER- tumors and dominant late-onset modes for HER2-ER+ breast cancers. Epidemiologic patterns for concordant HER2+ER+ (double-positive) and HER2-ER- (double-negative) were intermediate to discordant HER2+ER- and HER2-ER+. CONCLUSION: Results showed contrasting incidence patterns for HER2+ (HER2+ER-) and ER+ (HER2-ER+) breast cancers, diagnosed in 1995. Though sample sizes were small, this demonstration project validates the potential utility of the RTR for supplementing the SEER program.


Subject(s)
Breast Neoplasms/genetics , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Age Distribution , Age of Onset , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Female , Humans , Immunohistochemistry , Incidence , Middle Aged , Oligonucleotide Array Sequence Analysis , Receptors, Progesterone/analysis , Registries , Reproducibility of Results , SEER Program
5.
Endocr Relat Cancer ; 11(2): 191-205, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15163298

ABSTRACT

The ras family of proto-oncogenes are upstream mediators of several essential cellular signal transduction pathways involved in cell proliferation and survival. Point mutations of ras oncogenes result in constitutively active Ras and have been shown to be oncogenic. However, ras activation can occur in the absence of ras mutations secondary to upstream receptor activation. The first important step in Ras activation is farnesylation by farnesyl transferase, and inhibitors of this enzyme have been demonstrated to inhibit Ras signaling, and have anti-tumor effects. However, it is now clear that farnesyl transferase inhibitors (FTIs) have activity independent of Ras, most likely due to effects on prenylated proteins downstream of Ras, which explains their activity in several malignancies, including breast cancer, where ras mutations are rare. Several FTIs are in clinical development for the treatment of solid tumors. Preclinical evidence suggests that FTIs can inhibit breast cancers in vitro and in vivo, and a phase II trial of the FTI, R115777, in patients with advanced breast cancer produced encouraging results. Based on prior successful outcomes with agents targeting the estrogen and epidermal growth factor receptor pathways in breast cancer, the FTIs, used alone or more likely with other agents, may be the next exciting targeted therapy in breast cancer.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Alkyl and Aryl Transferases/metabolism , Animals , Breast Neoplasms/enzymology , Farnesyltranstransferase , Female , Humans
7.
Clin Cancer Res ; 7(12): 4149-55, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751515

ABSTRACT

PURPOSE: Arzoxifene (Arzox) is a novel benzothiophene analogue with selective estrogen receptor modulator activity similar to raloxifene. Arzox is being developed as a treatment for breast cancer and has a predominantly antiestrogenic effect on the rodent uterus. Our objectives were to verify whether the novel selective estrogen receptor modulator, Arzox, can be a good first-line agent and also be effective at controlling the growth of endometrial cancer after exposure to tamoxifen (Tam). EXPERIMENTAL DESIGN: We compared the effects of Tam and Arzox on the growth of estrogen responsive ECC-1 endometrial cancer cells in vitro, and we determined their antitumor effects on ECC-1 and EnCa101 endometrial carcinoma growth in athymic mice. RESULTS: We observed that estrogen receptor protein expression is down-regulated by Arzox to the same extent as raloxifene, whereas 4-hydroxytamoxifen, the active metabolite of Tam, does not affect estrogen receptor protein levels. Tam and Arzox inhibit the growth of Tam-naïve ECC-1 tumors in athymic mice. However when Tam-stimulated or estrogen-stimulated (which had been treated with Tam previously) EnCa101 endometrial tumors were treated with Tam or Arzox, we observed a stimulatory effect of both compounds in these models. CONCLUSIONS: The results indicate that Arzox may be a good first-line agent, but it may be ineffective at controlling the growth of endometrial cancer after exposure to Tam. Our data suggest that Arzox stimulates endometrial tumor growth to at least the same extent as Tam, thereby suggesting a limited role as a second-line agent for the patient on Tam who develops occult endometrial cancer.


Subject(s)
Endometrial Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Piperidines/therapeutic use , Tamoxifen/analogs & derivatives , Thiophenes/therapeutic use , Animals , Cell Division/drug effects , Down-Regulation/drug effects , Endometrial Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Receptors, Estrogen/genetics , Tamoxifen/therapeutic use , Transplantation, Heterologous , Tumor Cells, Cultured
8.
J Physiol ; 537(Pt 1): 251-65, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11711578

ABSTRACT

1. We examined the upper airway (UA) motor response to upper airway negative pressure (UANP) in the rat. We hypothesized that this response is mediated by superior laryngeal nerve (SLN) afferents and is not confined to airway dilator muscles but also involves an increase in motor drive to tongue retractor and pharyngeal constrictor muscles, reflecting a role for these muscles in stabilizing the UA. 2. Experiments were performed in 49 chloralose-anaesthetized, tracheostomized rats. Subatmospheric pressure in the range 0 to -30 cmH(2)O was applied to the isolated UA. Motor activity was recorded in separate experiments from the main trunk of the hypoglossal nerve (XII, n = 8), the pharyngeal branch of the glossopharyngeal nerve (n = 8), the medial and lateral branches of the XII (n = 8) and the pharyngeal branch of the vagus (n = 8). Afferent activity was recorded from the whole SLN in six experiments. 3. All UA motor outflows exhibited phasic inspiratory activity and this was significantly (P < 0.05) increased by UANP. Tonic end-expiratory activity increased significantly in response to pressures more negative than -20 cmH(2)O. Bilateral section of the SLN also increased (P < 0.05) motor activity and abolished the responses to UANP. Electrical stimulation of the SLN inhibited inspiratory XII activity. SLN afferents were tonically active and inhibited by UANP. 4. We conclude that UANP causes a reflex increase in motor drive to pharyngeal dilator, tongue retractor and pharyngeal constrictor muscles via afferent fibres in the SLN. Tonic activity in SLN afferent fibres at zero transmural pressure exerts a marked inhibitory effect on UA motor outflow.


Subject(s)
Nasal Cavity/physiology , Reflex/physiology , Respiratory Physiological Phenomena , Trachea/physiology , Afferent Pathways/physiology , Animals , Electric Stimulation , Hypoglossal Nerve/physiology , Laryngeal Nerves/physiology , Lung/physiology , Lung Volume Measurements , Male , Pharynx/innervation , Phrenic Nerve/physiology , Pressure , Rats , Rats, Sprague-Dawley , Respiration , Respiratory Muscles/physiology , Vagus Nerve/physiology
9.
Oncology (Williston Park) ; 15(9): 1177-85, 1189-90; discussion 1190-4, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11589065

ABSTRACT

Tamoxifen (Nolvadex), a selective estrogen-receptor modulator, or SERM, is currently the endocrine therapy of choice for all stages of hormone-responsive breast cancer. Only tamoxifen has been approved by the US Food and Drug Administration to reduce the incidence of breast cancer in high-risk women. Despite tamoxifen's antiestrogenic effects in breast tissue, it exhibits paradoxical estrogenic effects in other tissues in the body. These effects result in the maintenance of bone mineral density, but a three- to fourfold increase in endometrial cancer in postmenopausal women. Additionally, tamoxifen can result in troublesome hot flashes and serious thromboembolic events. For this reason, current research is focusing on new agents that may maintain the beneficial effects of tamoxifen while reducing its adverse effects. Raloxifene (Evista) is another SERM, approved for the prevention of osteoporosis in postmenopausal women and now being compared with tamoxifen in an ongoing breast cancer prevention trial. Like tamoxifen, raloxifene is associated with hot flashes and thromboembolic events, but its association with the risk of endometrial cancer is unknown. A number of new SERMs are in preclinical or clinical development in an attempt to improve upon the safety profile of tamoxifen. Additionally, selective aromatase inhibitors are being examined in the early breast cancer setting.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Neoplasms, Hormone-Dependent/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Breast Neoplasms/prevention & control , Clinical Trials as Topic , Endometrial Neoplasms/chemically induced , Estrogen Antagonists/adverse effects , Female , Humans , Middle Aged , Neoplasms, Hormone-Dependent/prevention & control , Pilot Projects , Raloxifene Hydrochloride/adverse effects , Raloxifene Hydrochloride/therapeutic use , Tamoxifen/adverse effects , Venous Thrombosis/chemically induced
10.
Exp Physiol ; 86(5): 641-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11571493

ABSTRACT

The effects of carbonic anhydrase inhibition on the responsiveness to CO2 of pressure-sensitive laryngeal receptors were examined in anaesthetised, paralysed cats. Laryngeal CO2-sensitive receptors from the superior laryngeal nerve were selected by their responsiveness to intralaryngeal pressure and to perfusion of solution equilibrated with 9% CO2. The carbonic anhydrase inhibitor, methazolamide, when given intralaryngeally at 10(-4) M, diminished or abolished the responses to the CO2-equilibrated solution in four of six pressure-sensitive receptors. Histochemical staining for carbonic anhydrase activity showed that the larynges perfused with methazolamide had diminished carbonic anhydrase activity, especially on the superficial layers of surface epithelium. Compared to untreated controls, when given intravenously (50 mg x kg(-1)) methazolamide diminished or abolished the responses to CO2 of five of the six fibres studied. Histochemical staining of these larynges showed no carbonic anhydrase reactivity at the sites of laryngeal receptors. These results suggest that the responses to CO2 of laryngeal pressure-sensitive receptors are dependent on the presence of carbonic anhydrase. Inhibition of laryngeal receptor carbonic anhydrase activity by methazolamide is more reliably achieved by systemic rather than by luminal administration.


Subject(s)
Carbon Dioxide/metabolism , Carbonic Anhydrases/metabolism , Chemoreceptor Cells/metabolism , Laryngeal Nerves/metabolism , Larynx/physiology , Animals , Carbonic Anhydrase Inhibitors/pharmacology , Cats , Electrophysiology , Female , Humans , Immunohistochemistry , Larynx/cytology , Male , Mechanoreceptors/metabolism , Methazolamide/pharmacology , Pressure
11.
Clin Cancer Res ; 7(8): 2505-12, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489833

ABSTRACT

PURPOSE: Cross-resistance is the primary issue facing the evaluation of new antiestrogens to treat metastatic breast cancer because they may be tested, initially, in populations of patients that have failed long-term adjuvant tamoxifen (Tam) therapy. EXPERIMENTAL DESIGN: We have tested the benzothiophene derivatives, arzoxifene (Arzox; LY353381) and LY117018 in two models of Tam-stimulated tumor growth derived from either MCF-7 (M. M. Gottardis and V. C. Jordan, Cancer Res., 48: 5183-5187, 1988) or T47D (J. MacGregor Schafer et al., Clin. Cancer Res., 6: 4373-4380, 2000) breast cancer cells. RESULTS: Using the MCF-7:Tam model, we found that both Arzox and LY117018 (1.5 mg/day) resulted in tumor growth and, therefore, were partially cross-resistant with Tam. Next, using the T47D:17beta-estradiol (E(2)) model, we compared the antiestrogenic/antitumor properties of Arzox and LY117018 and determined that neither Arzox nor LY117018 caused T47D:E(2) tumor growth after 21 weeks. In addition, we determined that long-term treatment does not result in failure and subsequent development of transplantable Arzox- or LY117018-stimulated tumors. To establish whether Arzox and LY117018 are cross-resistant in T47D:Tam tumors, mice were treated with Arzox or LY117018 (1.5 mg/day), and, again, we found that neither resulted in the growth of transplantable tumors. Lastly, we showed that Arzox and LY117018 were only partially able to compete with postmenopausal E(2) (0.3 cm silastic capsule) in T47D:Tam tumors. However, when T47D:E(2) tumors were treated for 7 days instead of 5 days, both Arzox and LY117018 were more effective. CONCLUSIONS: Arzox is not cross-resistant with Tam in the T47D athymic mouse model but does exhibit cross-resistance in the MCF-7 model.


Subject(s)
Breast Neoplasms/drug therapy , Piperidines/pharmacology , Pyrrolidines/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/therapeutic use , Thiophenes/pharmacology , Animals , Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Estradiol/therapeutic use , Female , Humans , Mice , Mice, Nude , Time Factors , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Semin Oncol ; 28(3): 260-73, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11402436

ABSTRACT

Tamoxifen, a selective estrogen receptor modulator (SERM), is the treatment of choice for all stages of hormone-responsive breast cancer and has been shown to prevent breast cancer in high-risk women. Despite acting as an antiestrogen on the breast, tamoxifen has partial estrogenic effects on other target tissues. These partial estrogen agonistic actions produce beneficial effects on bones and the lipid profile in postmenopausal women. However, tamoxifen is associated with an increase in endometrial cancer. Additionally, its antiestrogenic effects in the central nervous system result in hot flashes in postmenopausal women. Raloxifene is another SERM approved for the prevention of osteoporosis in postmenopausal women. Like tamoxifen, raloxifene appears to prevent breast cancer in high-risk women and has not, to date, been noted to increase the incidence of endometrial cancer. The Study of Tamoxifen and Raloxifene will compare the effects of the two agents on breast cancer prevention and endometrial cancer risk. A number of new agents are being developed for breast cancer treatment and prevention and osteoporosis prevention. These include other SERMs, selective estrogen receptor downregulators (SERDs), and aromatase inhibitors. It is hoped that one of these new agents will be the ideal agent, acting as an antiestrogen on breast and endometrium while having estrogenic effects on bones, the lipid profile, and the central nervous system. Semin Oncol 28:260-273.


Subject(s)
Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Raloxifene Hydrochloride/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Aromatase Inhibitors , Breast Neoplasms/prevention & control , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Estrogen Antagonists/therapeutic use , Female , Humans , Neoplasms, Hormone-Dependent/prevention & control , Raloxifene Hydrochloride/chemistry , Raloxifene Hydrochloride/pharmacology , Tamoxifen/analogs & derivatives , Tamoxifen/chemistry , Tamoxifen/pharmacology
15.
Clin Cancer Res ; 6(11): 4373-80, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11106256

ABSTRACT

MCF-7 cells are used routinely to study tamoxifen-stimulated drug resistance in vivo. However, unlike MCF-7 cells, T47D cells express mutant p53 protein and lose the estrogen receptor (ER) during long-term estrogen deprivation in vitro [Pink et al., Br. J. Cancer, 74: 1227-1236, 1996 (erratum, Br. J. Cancer, 75: 1557, 1997)]. As a result, T47D tumors may respond differently from MCF-7 tumors to long-term tamoxifen treatment. Ovariectomized athymic mice were given injections bilaterally with T47D cells (5 x 10(5)) into the mammary fat pads. A rapidly growing estradiol responsive tumor (T47D:E2) was established and 0.5 mg of tamoxifen given daily blocked estrogen-stimulated growth. In subsequent experiments, low doses of tamoxifen (0.17 mg or 0.5 mg) did not produce tamoxifen-stimulated tumors at 14 weeks, whereas high-dose tamoxifen (1.5 mg) consistently produced tamoxifen-stimulated tumors (T47D:Tam; 17 tumors/20 sites) at 8 weeks. In contrast, 1.5 mg of tamoxifen produced tamoxifen-stimulated MCF-7 tumors (MCF-7: Tam2) at a slower rate (20 weeks) and less consistently (14 tumors/26 sites). When the T47D:Tam tumor was passaged, it grew maximally with either 1.5 mg of tamoxifen or a 1-cm estradiol (premenopausal levels) capsule, and similar results were obtained with MCF-7:Tam2 tumors. Interestingly, when T47D:Tam tumors were treated with the 0.5 mg of tamoxifen, tumors grew only to 50% maximum. All of the tumors originating from MCF-7 and T47D cells expressed ER at similar levels; therefore, tamoxifen did not select for an ER-negative tumor. In conclusion, we have shown that tamoxifen-stimulated T47D p53 mutant tumors can be developed rapidly with high-dose therapy (1.5 mg daily). The results from this model provide new opportunities to investigate the rapid development of drug resistance to adjuvant tamoxifen in patients with mutant p53 breast tumors.


Subject(s)
Estrogen Receptor Modulators/pharmacology , Genes, p53 , Mammary Neoplasms, Experimental/drug therapy , Tamoxifen/pharmacology , Animals , Drug Resistance, Neoplasm , Endothelial Growth Factors/analysis , Estrogen Receptor alpha , Female , Humans , Lymphokines/analysis , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Receptors, Estrogen/analysis , Transplantation, Heterologous , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Cancer Treat Res ; 103: 183-207, 2000.
Article in English | MEDLINE | ID: mdl-10948447

Subject(s)
Anticarcinogenic Agents/therapeutic use , Breast Neoplasms/prevention & control , Estrogens , Neoplasms, Hormone-Dependent/prevention & control , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Adult , Aged , Animals , Anticarcinogenic Agents/administration & dosage , Bone and Bones/drug effects , Breast Diseases/epidemiology , Breast Neoplasms/epidemiology , Breast Neoplasms/therapy , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Chemotherapy, Adjuvant , Clinical Trials as Topic , Endometrial Neoplasms/chemically induced , Endometrial Neoplasms/epidemiology , Female , Gonadal Steroid Hormones/analysis , Humans , Lipids/blood , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred C3H , Middle Aged , Multicenter Studies as Topic , Neoplasms, Hormone-Dependent/epidemiology , Neoplasms, Second Primary/epidemiology , Neoplastic Syndromes, Hereditary/epidemiology , Osteoporosis, Postmenopausal/prevention & control , Pilot Projects , Precancerous Conditions/epidemiology , Premenopause , Prospective Studies , Raloxifene Hydrochloride/administration & dosage , Raloxifene Hydrochloride/adverse effects , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Randomized Controlled Trials as Topic , Receptors, Estrogen/drug effects , Reproductive History , Risk Factors , Safety , Selective Estrogen Receptor Modulators/administration & dosage , Selective Estrogen Receptor Modulators/adverse effects , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/administration & dosage , Tamoxifen/adverse effects , Tamoxifen/pharmacology
17.
Respir Physiol ; 121(1): 65-74, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10854624

ABSTRACT

It has been demonstrated previously that isohydric hypercapnia (IH) does not affect agonist-induced tension development in pulmonary arteries. The aim of the present study was to examine the effects of IH on depolarisation-induced, steady state tension in the isolated rat pulmonary artery. Rings were submaximally contracted with high KCl under control conditions (5% CO(2)-95% air). IH was achieved by switching to a modified PSS (isosmotic substitution of NaHCO(3) for NaCl), equilibrated with 10% CO(2) in air. On switching to IH, a significant increase in mean (+/-SEM) tension (25.3+/-6.3% Tmax) was observed in endothelium intact rings (n=6). Endothelial removal significantly reduced this response. Non-specific inhibition of nitric oxide synthase (NOS) isoenzymes (L-NAME, 10(-3) M) abolished the IH-induced increase in tension while inhibition of neuronal NOS (TRIM, 10(-5) M) was without effect. The relaxant response to the nitric oxide donor sodium nitroprusside was similar in IH and control conditions. These results suggest that IH caused an endothelium-dependent increase in depolarisation-induced tension by reducing NO production.


Subject(s)
Endothelium, Vascular/physiology , Hypercapnia/physiopathology , Pulmonary Artery/physiopathology , Acetylcholine/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , In Vitro Techniques , Male , Membrane Potentials/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Pulmonary Artery/drug effects , Rats , Rats, Sprague-Dawley
18.
Clin Cancer Res ; 6(5): 2028-36, 2000 May.
Article in English | MEDLINE | ID: mdl-10815929

ABSTRACT

The estrogen receptor (ER)-positive MCF-7 breast cancer cell line can be transplanted into athymic mice and grown into tumors with estradiol (E2) support. Tamoxifen (TAM) blocks E2-stimulated tumor growth; however, continuous TAM treatment results in transplantable tumors within a year that will grow with either E2 or TAM (M. M. Gottardis and V. C. Jordan, Cancer Res., 48: 5183-5187, 1988). Although this model may represent the development of TAM resistance for the treatment of advanced breast cancer, no laboratory model exists to study the exposure of breast cancer to 5 years of adjuvant TAM therapy. We have addressed this issue and report the development and characterization of two tumor lines, MCF-7TAM and MT2, which have been serially transplanted into TAM-treated athymic mice for >5 years. The MCF-7TAM tumor rapidly regresses in response to E2 and then about 50% of tumors regrow in response to E2. Interestingly, tumor regression does not occur if TAM treatment is stopped, probably because E2 levels are too low in ovariectomized athymic mice. The development of the antitumor effect of E2 was documented for MT2 tumors over a 1-year period; TAM-stimulated tumor growth was retained, but E2 caused progressively less of a stimulatory effect. Most importantly, E2-stimulated tumors that regrew after initial tumor regression in both MCF-7TAM and MT2 lines were again responsive to TAM to block E2-stimulated growth. Unlike MCF-7 tumors, the MT2 tumor line contains a single point mutation, Asp351Tyr, in the ER, which was retained after the development of E2-stimulated regrowth. The mutation is associated with increased estrogen-like actions for the TAM-ER complex (A. S. Levenson et al., Br. J. Cancer, 77: 1812-1819, 1998), but we conclude that the mutant ER is not required for TAM resistance. On the basis of the new breast cancer models presented, we propose a cyclic sensitivity to TAM that may have important clinical implications: (a) it is possible that a woman's own estrogen may produce an antitumor effect on the presensitized micrometastatic disease after 5 years of TAM. Long-term antitumor action occurs because the drug is stopped, but resistance accumulates and tumors start to grow if adjuvant therapy is continued; and (b) although in the clinic TAM-resistant tumors respond to second-line therapies that cause estrogen withdrawal, e.g., pure antiestrogens or aromatase inhibitors, estrogen therapy may also be effective and return the tumor to TAM responsiveness. In this way, a hormone-responsive tumor may be controlled longer in the patient with advanced disease.


Subject(s)
Antineoplastic Agents, Hormonal/adverse effects , Estradiol/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Tamoxifen/adverse effects , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Cell Division/drug effects , Dose-Response Relationship, Drug , Female , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Neoplasm Transplantation , Receptors, Estrogen/genetics , Tamoxifen/therapeutic use , Time Factors , Transplantation, Heterologous , Tumor Cells, Cultured
19.
Am J Physiol Heart Circ Physiol ; 278(2): H331-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666061

ABSTRACT

Chronic hypercapnia is commonly found in patients with severe hypoxic lung disease and is associated with a greater elevation of pulmonary arterial pressure than that due to hypoxia alone. We hypothesized that hypercapnia worsens hypoxic pulmonary hypertension by augmenting pulmonary vascular remodeling and hypoxic pulmonary vasoconstriction (HPV). Rats were exposed to chronic hypoxia [inspiratory O(2) fraction (FI(O(2))) = 0.10], chronic hypercapnia (inspiratory CO(2) fraction = 0.10), hypoxia-hypercapnia (FI(O(2)) = 0.10, inspiratory CO(2) fraction = 0.10), or room air. After 1 and 3 wk of exposure, muscularization of resistance blood vessels and hypoxia-induced hematocrit elevation were significantly inhibited in hypoxia-hypercapnia compared with hypoxia alone (P < 0.001, ANOVA). Right ventricular hypertrophy was reduced in hypoxia-hypercapnia compared with hypoxia at 3 wk (P < 0.001, ANOVA). In isolated, ventilated, blood-perfused lungs, basal pulmonary arterial pressure after 1 wk of exposure to hypoxia (20.1 +/- 1.8 mmHg) was significantly (P < 0.01, ANOVA) elevated compared with control conditions (12.1 +/- 0.1 mmHg) but was not altered in hypoxia-hypercapnia (13.5 +/- 0.9 mmHg) or hypercapnia (11.8 +/- 1.3 mmHg). HPV (FI(O(2)) = 0.03) was attenuated in hypoxia, hypoxia-hypercapnia, and hypercapnia compared with control (P < 0.05, ANOVA). Addition of N(omega)-nitro-L-arginine methyl ester (10(-4) M), which augmented HPV in control, hypoxia, and hypercapnia, significantly reduced HPV in hypoxia-hypercapnia. Chronic hypoxia caused impaired endothelium-dependent relaxation in isolated pulmonary arteries, but coexistent hypercapnia partially protected against this effect. These findings suggest that coexistent hypercapnia inhibits hypoxia-induced pulmonary vascular remodeling and right ventricular hypertrophy, reduces HPV, and protects against hypoxia-induced impairment of endothelial function.


Subject(s)
Hypercapnia/physiopathology , Hypoxia/physiopathology , Pulmonary Circulation , Animals , Blood Pressure , Blood Vessels/pathology , Blood Vessels/physiopathology , Chronic Disease , Endothelium, Vascular/physiopathology , Hematocrit , Hypercapnia/blood , Hypertrophy, Right Ventricular/etiology , Hypoxia/blood , In Vitro Techniques , Male , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Vasoconstriction , Vasodilation
20.
Clin Cancer Res ; 6(12): 4893-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11156249

ABSTRACT

The triphenylethylene antiestrogens, idoxifene (Idox) and toremifene (Tor), are structurally related analogues of tamoxifen (Tam) and were developed to improve the therapeutic index for advanced breast cancer patients. However, the issue of cross-resistance with Tam for these new agents is critical for clinical testing because the majority of breast cancer patients have already received or failed adjuvant Tam. The goal of this study was to determine the effectiveness of Idox as an antitumor agent in three models of Tam-stimulated breast cancer in athymic mice and compare the results with the actions of Tor and ICI 182,780 in a Tam-stimulated MCF-7 tumor model. We first compared the activities of Tam and Idox in the 17beta-estradiol (E2)-stimulated MCF-7 tumor line MT2:E2. Tam and Idox reduced E2-stimulated growth by 65-70% (week 9: P = 0.0009 for Tam, P = 0.0005 for Idox versus E2 alone). However, Tam (1.5 mg daily) and Idox (1.0 mg daily) both produced T47D breast tumors in athymic mice during 23 weeks of treatment (12 tumors/22 sites and 15 tumors/18 sites, respectively). Tam and Idox stimulated tumor growth equally in two different Tam-stimulated MCF-7 models and in a T47D model. Tor was completely cross-resistant with Tam in the MCF-7 tumor model, which implied that neither Idox nor Tor would be effective as a second-line endocrine therapy after Tam failure and may offer no therapeutic advantages over Tam as adjuvant therapies. In contrast, ICI 182,780, a pure antiestrogen currently being tested as a treatment for breast cancer after Tam failure, had no growth-stimulatory effect on the MCF-7 Tam-stimulated breast tumor line. This agent may provide an advantage as an adjuvant therapy and increase the time to treatment failure.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/chemistry , Estrogen Receptor Modulators/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Stilbenes/chemistry , Tamoxifen/analogs & derivatives , Tamoxifen/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Estradiol/metabolism , Female , Fulvestrant , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Tamoxifen/pharmacology , Time Factors , Toremifene/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL