Women and lung cancer: Epidemiology, tumor biology, and emerging trends in clinical research

Women and lung cancer: Epidemiology, tumor biology, and emerging trends in clinical research

Lung Cancer (2007) 55, 15—23 available at www.sciencedirect.com journal homepage: www.elsevier.com/locate/lungcan REVIEW Women and lung cancer: Ep...

178KB Sizes 0 Downloads 34 Views

Lung Cancer (2007) 55, 15—23

available at www.sciencedirect.com

journal homepage: www.elsevier.com/locate/lungcan

REVIEW

Women and lung cancer: Epidemiology, tumor biology, and emerging trends in clinical research Chandra P. Belani a, Sherry Marts b,∗, Joan Schiller c, Mark A. Socinski d a

University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States Society for Women’s Health Research, 1025 Connecticut Ave. NW, Suite 701, Washington, DC 20036, United States c Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical School, Dallas, TX, United States d Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States b

Received 14 July 2006; received in revised form 14 September 2006; accepted 18 September 2006

KEYWORDS Lung neoplasms; Female; Biomedical research; Therapy

Summary Lung cancer is the leading cause of cancer-related death in both men and women. Environmental carcinogens, particularly tobacco smoke, play a dominant role in the development of lung cancer, although 10—15% of all patients diagnosed are non-smokers. In addition, emerging data demonstrate sex-specific differences in lung cancer susceptibility and prognosis. This implies that the development of lung cancer is modulated by complex interactions between genetic, hormonal, behavioral, and environmental factors. A better understanding of the differences between men and women and their impact on the prevention, diagnosis, and treatment of lung cancer requires continued basic and clinical research. Recent data on the epidemiological aspects of lung cancer in women, lung tumor biology, and emerging trends in clinical research were presented at a thought leaders’ roundtable hosted by the Society for Women’s Health Research. The panel concluded that as the patient population in lung cancer is changing from mostly male smokers to include women and non-smokers, an urgent need exists to increase awareness and research funding to improve lung cancer care, particularly in women. To further improve survival in this disease, both clinical characteristics and tumor biology should be considered in the development of new treatment options. © 2006 Elsevier Ireland Ltd. All rights reserved.

Contents 1. 2.



Introduction............................................................................................................. Men, women, and health ................................................................................................

Corresponding author. Tel.: +1 202 496 5019; fax: +1 202 833 3472. E-mail address: [email protected] (S. Marts).

0169-5002/$ — see front matter © 2006 Elsevier Ireland Ltd. All rights reserved. doi:10.1016/j.lungcan.2006.09.008

16 16

16 3. 4. 5. 6.

C.P. Belani et al. Lung cancer: epidemiology and risk factors ............................................................................. Sex differences in lung tumor biology ................................................................................... Current and experimental treatment options............................................................................ Conclusion .............................................................................................................. Conflict of interest statement ........................................................................................... References ..............................................................................................................

1. Introduction Women’s healthcare issues related to breast cancer and gynecological malignancies have received substantial attention and research funding in the past two decades. However, differences between men and women in the development and prognosis of common tumor types, such as melanoma [1], colorectal [2], or lung cancer [3], indicate the need for broader sex-specific research to optimize treatment options for men and women. These differences in tumor etiology may be influenced by physiological factors, such as the role of sex hormones, or by behavioral and environmental factors, such as levels of sun exposure or smoking habits. Overall, the variability in tumor development is based on a complex interaction between genetic, hormonal, and behavioral factors. Lung cancer is the leading cause of cancer-related death in women. An estimated 73,000 women will die from progressive lung cancer in 2005, accounting for 27% of all cancer death in US women (compared to 15 and 6% of cancer deaths due to breast and ovarian cancer, respectively; ref. [4]). Increasing evidence suggests that the relative risk for lung cancer, the relationship between smoking and lung cancer, and response to therapy may be different for women and men. In February 2006, the Society for Women’s Health Research hosted a thought leaders’ roundtable to review issues and advances related to sex differences in the development and treatment of lung cancer. Recent data on the epidemiological aspects of lung cancer in women, lung tumor biology, and emerging trends in clinical research were presented. The discussions, summarized in this review, clearly highlight the need for basic and clinical research to translate information on differences in tumor biology and risk factors between men and women into preventative, diagnostic, and therapeutic practice to improve women’s healthcare.

2. Men, women, and health Historically, women have been underrepresented in clinical research. In 1977, the US Food and Drug Administration (FDA) restricted research on women of childbearing potential in reaction to the birth defects resulting from thalidomide and diethylstilbestrol administered during pregnancy. As a result, findings from clinical studies on men were extrapolated to female patients, not taking into account potential differences in pharmacokinetics, pharmacodynamics, and safety between the sexes [5,6]. In addition, drugs that are potentially beneficial to women may have been eliminated in early phases of clinical testing when the test group did not include women and no benefits were manifest in male subjects [7]. In 1993, the FDA published

16 18 19 20 20 21

new guidelines encouraging participation of women in clinical studies and also requiring study sponsors to collect and analyze data separately by sex to identify any differences in drug response and toxicity. However, women are still underrepresented in cancer trials [8,9], and for most drugs currently on the market, prospective evaluation of efficacy and toxicity by sex is not available. Sex clearly affects health as men and women have different patterns and presentations of an illness, as well as statistically different life-spans [10,11]. The phenotypic differences between males and females are determined, initially, by genes on the sex chromosomes. Male sex is initiated and determined by the presence of the SRY-gene on the Y-chromosome. The expression of these differences in the context of physiology and endocrine function will vary throughout childhood, adolescence, adulthood, and post-menopause. On average, women, relative to men, have a higher percentage of body fat, smaller muscle mass, lower blood pressure, higher levels of estrogens and progestins, and lower levels of androgen (reviewed in ref. [5]). In addition, women, but not men, undergo hormonal fluctuations associated with reproduction (such as the ovarian cycle and pregnancy) that influence numerous body systems. During menopause, reduced estrogen levels affect not only the ovaries, uterus, and breast, but also other tissues such as the brain, skin, cardiovascular tissue, and bone [12]. Consequently, sex should be considered an important variable in research design, both in basic and clinical research as well as epidemiology. Clinical research should also take into consideration that differences between men and women may vary with age and endocrine status. When reporting research findings in the medical literature, analyses by sex and endocrine status should be included whenever possible, even in the absence of significant differences in outcome between the sexes. A clearer understanding of the function of sex hormones in body systems other than the reproductive organs will allow for more individualized options for the prevention, diagnosis, and treatment of disease. A growing awareness and knowledge of biological sex differences will have an impact on healthcare and health policy, and improve outcome for both men and women.

3. Lung cancer: epidemiology and risk factors In 2005, 172,570 patients (93,010 men and 79,560 women) were diagnosed with lung cancer, and 163,510 patients (90,490 men and 73,020 women) were expected to die of this disease [4]. Because lung cancer is asymptomatic in its early stages, only 18% of cases are still confined to the primary site at diagnosis (local disease); 38% are diagnosed after the cancer has spread to regional lymph-nodes or directly beyond

Lung cancer in women Table 1A

17

5-year survival rates in men and women by stage in NSCLC and SCLC [13] NSCLC

All stages Local disease Regional disease Metastatic disease

Men (%)

Women (%)

Men (%)

Women (%)

14.9 46.6 15.9 2.0

19.6 56.0 18.5 2.3

5.6 20.7 9.9 1.8

7.2 21.6 11.7 2.2

the primary site (regional disease), and 37% are diagnosed after the cancer has already spread to other parts of the body (metastatic disease). The corresponding 5-year relative survival rates are 51.3% for localized, 17.1% for regional, and 2.1% for metastatic disease [13]. Women have a better relative survival compared to men for each stage of the disease (Table 1A; ref. [14]), and male sex is a consistently unfavorable prognostic indicator in advanced disease (Table 1B; refs. [15—20]). In women, smoking status prior to lung cancer diagnosis and duration of smoking abstinence are predictive of survival: risk of death was higher for current or past female smokers compared to never-smokers, a trend that was not observed in men [21,22]. Overall, women diagnosed with lung cancer are more likely to be non-smokers than male patients, and women are also overrepresented among younger patients [23—26]. Lung cancer historically has been more prevalent in men than women; however, the male/female incidence ratio has narrowed dramatically, from 3.65 in 1975 to 1.65 in 1999, as the incidence rate in men declined while the rate in women continued to rise slowly [14]. This rising incidence of lung cancer in women is primarily due to an increase in tobacco use which started in the 1940s. The lung cancer death rate in women has subsequently increased rapidly since the 1960s, from about 5 cases per 100,000 women to an estimated 40 per 100,000 in 2000. Today, lung cancer is the most common cause of cancer death in women (27%), claiming more lives than breast and colorectal cancer combined (15 and 10%, respectively; ref. [4]). Reducing the incidence of lung cancer death in women requires renewed efforts towards smoking cessation and prevention. Although smoking rates in women have dropped since the 1960s, 19% of women ≥18 years old were current smokers in 2004, compared to 23% of men [27]. Moreover, a near equal prevalence of cigarette smoking has been reported in male and female adolescents [27,28]. Factors associated with the initiation of cigarette smoking in young adults vary between the sexes; particularly, in young

Table 1B

SCLC

women, vulnerability to depression and weight concerns are associated with an increased risk of smoking initiation [29,30]. Future intervention programs may be able to specifically target young women at increased risk for smoking initiation or continuation. Lung cancer histologies include squamous cell carcinoma, adenocarcinoma, small cell carcinoma, and large cell carcinoma. Non-small cell lung cancers (NSCLC) account for approximately 85% of lung cancer, and include the histological subtypes squamous cell carcinoma and large cell carcinoma, which arise from epithelial cells lining the bronchi, and adenocarcinoma, which develops from glandular tissue in the peripheral regions of the lung. The proportional occurrence of these histological subtypes differs significantly between men and women [14]. Adenocarcinoma is currently the most common histological subtype in both men and women, and women have proportionally more adenocarcinoma and less squamous cell carcinoma compared to men (Table 2A). Cigarette smoking has been linked to all four histological subtypes; however, the proportion of non-smokers is highest in those who develop adenocarcinoma (Table 2B; ref. [33]). The incidence rates for the various histological subtypes have changed over time and reflect changes in smoking habits. For example, incidence rates for squamous cell carcinoma decreased in men between 1975 and 1999 while increasing slightly in women. On the other hand, the incidence rate of adenocarcinoma increased in both in men and women during that same time period, with a greater increase observed in women [14,34]. The increase in adenocarcinoma has been associated with the introduction of low-tar cigarettes that enhance delivery of smoke to peripheral regions of the lungs [35]. While smoking is major risk factor, only a minority of smokers develop lung cancer. In addition, approximately 10—15% of patients diagnosed are non-smokers. The ratio of women to men in patients with lung cancer who have never smoked is approximately 3:1 [24,36]. The risk for

Male sex is a poor prognostic factor in advanced lung cancer

O’Connell et al. [15] Ferguson et al. [16] Shinkai et al. [17] Albain et al. [18] Paesmans et al. [19] Johnson et al. [20]

N

%Female

Hazard ratio

P-value

378 299 192 2531 1052 378

30 45 29 23 10 28

0.71 0.75 0.62 0.77 0.70 0.77

0.001 0.044 0.02 <0.0001 0.03 0.002

18 Table 2A

C.P. Belani et al. Proportion of patients diagnosed with either squamous cell carcinoma or adenocarcinoma by sex Adenocarcinoma

Moore et al. [31] Fu et al. [14] Visbal et al. [24] Chen et al. [32] Ringer et al. [26]

Squamous cell carcinoma

Men (%)

Women (%)

Men (%)

Women (%)

40 33 48 42 38

50 45 59 73 47

30 30 32 41 38

20 21 22 16 24

developing lung cancer in both smokers and non-smokers is modulated by determinants that may vary between individuals. For example, data from the Environmental Protection Agency showed that environmental tobacco smoke accounts for approximately 3000 lung cancer deaths in the US each year among non-smokers [37]. The development of lung cancer despite the low level of exposure suggests that a subset of the general population may be more susceptible to the carcinogenic effects of tobacco smoke. Susceptibility is likely to be determined by each individual’s capacity to activate and detoxify carcinogens in tobacco smoke. Certain genetic polymorphisms in genes involved in the metabolism of carcinogens have been associated with an increased risk of lung cancer in smokers and never-smokers [38—40]. Further evidence for genetic susceptibility comes from studies on family history of lung cancer. A positive family history has been defined as a risk factor in non-smokers; particularly in the development of adenocarcinoma, in women, and in cases with an earlier age at onset [41—44]. Finally, an epidemiologic link between estrogen exposure and incidence of NSCLC has been suggested in Japanese non-smoking women [45]. Younger, presumably pre-menopausal, women appear to have shorter survival than older women: in an analysis of patients with advanced NSCLC enrolled in SWOG trials, women over 70 had a 34% 1-year survival compared with 11% for those under 45 [18]. Clearly, the multifactorial nature of the disease requires an improved understanding of the interplay between sex, environmental factors, and genetics [46,47].

4. Sex differences in lung tumor biology The available epidemiological data clearly demonstrate sexspecific differences in lung cancer susceptibility and prognosis. Women appear to have an increased susceptibility to

tobacco carcinogens, but have a lower rate of fatal outcome of lung cancer compared to men [48]. A biological explanation for these epidemiological findings is beginning to emerge as the comparison of lung tumors from male and female patients has identified various tumor characteristics that may affect tumor etiology. Smoking-related lung cancer is induced by the formation of DNA adducts in lung epithelial cells [49]. Processing of these adducts by the cellular DNA repair machinery may lead to mutations in genes that initiate or facilitate tumor growth, such as the tumor suppressor gene p53. The formation of DNA adducts is dependent on the enzymatic activation of several tobacco-related pro-carcinogens, including polycyclic aromatic hydrocarbon (PAH; ref. [50]). PAH activation is catalyzed by the cytochrome P450 enzymes CYP1A1 and CYP1B1, and inactivated by glutathione S-transferases (GSTs). The expression of CYP1A1 and CYP1B1 in lung tissue is significantly higher in current smokers compared to former and never-smokers [51]. Levels of lung DNA adducts correlate with the level of CYP1A1 expression. Among smokers, female patients had a 3.9-fold higher median level of CYP1A1 compared to males. In non-smokers, certain CYP1A1 polymorphisms are associated with an increased lung cancer risk, and this association can be modulated by ETS and genetic variations of GSTs [38,52,53]. Independent of smoking history, the combined variant genotype of CYP1A1 and GSTM1 contribute to an increased risk of lung cancer in women compared to men (odds ratio: 6.54 versus 2.36; ref. [54]). The carcinogens present in tobacco smoke are associated with specific mutations at a relatively small number of codons of tumor suppressor p53; active cigarette smoking is positively correlated with the total frequency of p53 mutations [55]. The most common p53 mutations in lung cancer are G:C to T:A transversions, a type of mutation rarely found in other tumor types. In contrast, transversions from G:C to

Table 2B Proportion of patients diagnosed with lung cancer by sex, histological subtype, and smoking status (based on Muscat and Wynder; ref. [33]) Men

Adenocarcinoma Squamous cell carcinoma Small cell carcinoma Other

Women

Current smokers (%)

Former smokers (%)

Never-smokers (%)

Current smokers (%)

Former smokers (%)

Never-smokers (%)

32 35 15 18

34 37 11 18

58 19 0 23

42 20 19 19

44 20 12 24

59 12 3 26

Lung cancer in women Table 3

19

Differences in treatment modality by sex

Surgery only Moore et al. [31] Radzikowska et al. [101] Fu et al. [14] Local Regional Radiotherapy only Moore et al. [31] Radzikowska et al. [101] Fu et al. [14] Local Regional

Women (%)

Men (%)

P-value

Survival

27.4 23.3

24.7 18.8

.031 .0001

Median: 55.4 months vs. 37.2 months (P < .001) NA

63.7 23.4

56.4 22.2

<.0001

2-year: 74.3% vs. 66.0% (P < .0001)

17.7 14.5

19.6 14.6

.031 NS

Median: 10.1 months vs. 8.0 months (P = .004) NA

18.2 39.4

23.4 42.5

<.0001

2-year: 12.2% vs. 15.5% (P < .0001)

A:T are more common in lung tumors from non-smokers and other types of cancer [56]. The prevalence of G:C to T:A transversions is approximately 30% in smokers, compared to 10% of non-smokers. The prevalence of G:C to A:T transversion is approximately 28% in smokers and 50% in non-smokers [55—57]. Analysis by sex shows that the tobacco-related p53 mutations are more common in women than men, evidence that women may be more susceptible to the carcinogenic effects of tobacco smoke [56—59]. In addition, the G:C to T:A transversion rate is significantly lower, and the G:C to A:T transversion significantly higher, in female nonsmokers compared to female smokers [57]. The dramatic shift in the p53 mutational spectrum between women who are non-smokers and those who smoke suggests that alternate tumorigenic pathways may be activated depending on exposure to tobacco smoke. If so, tumors from non-smoking women may be biologically different than those from male or female smokers. The heterogeneous nature of lung cancer first became evident when clinical responses to gefitinib, a tyrosine kinase inhibitor that targets the epidermal growth factor receptor (EGFR), were noted in patients with a mutated form of EGFR. Mutations in the tyrosine kinase domain of EGFR constitutively activate downstream signaling in the absence of ligand binding, and are associated with cellular proliferation and resistance to apoptosis. In lung cancers, these activating mutations are more common in women and non-smokers [60—63]. Mutations in K-RAS, a downstream signaling molecule in the EGFR pathway, may also be more common in women, but are significantly associated with smoking and levels of DNA adduct formation [64,65]. In hormone-dependent tissues, such as breast and ovary, estrogen regulates the transcription of target genes through interaction with estrogen receptors (ERs) alpha or beta. While important for normal tissue physiology, estrogen receptor-mediated signaling also promotes tumorigenesis, either in response to estrogen-binding or independent of estrogen through the expression of growth factors, particularly EGFR [66—70]. Similarly, estrogen plays a role in both normal pulmonary physiology and in the biology of non-small cell lung carcinoma (NSCLC) [71—74]. In vitro studies confirm that NSCLC cells respond to estrogens and anti-estrogens

by altering endogenous gene expression [75]. ER-beta, and to a lesser extent ER-alpha, are expressed in lung tumors from both men and women. Expression of ER-beta is associated with improved survival, while expression of ER-alpha is a poor prognostic factor [68,73,76,77]. In a large series of surgically resected NSCLC tumors, ER-beta was detected in 45.8% of cases. Overexpression of ER-beta was significantly more common in tumors from non-smokers (53.5%) than smokers (36.6%, P < .004). Among non-smokers, higher ERbeta expression was observed significantly more frequently in female patients (58.3%) than in male patients (40.9%) [73]. Current data related to tumor biology have started to provide an explanation for the risk factors associated with the development of lung cancer. Particularly, lung tumorigenesis may progress via different pathways in men and women depending on the metabolisms of tobacco-related carcinogen, molecular abnormalities, and hormonal status. A better understanding of the biological and molecular aspects contributing to these differences may allow the development of more effective therapy specifically tailored for men or women.

5. Current and experimental treatment options Initial treatment for patients with localized, early stage NSCLC typically includes surgical resection; patients with limited pulmonary reserve are candidates for radiotherapy. The treatment modality for early stage disease differs significantly between men and women (Table 3). In general, a higher proportion of women undergo surgery as part of a treatment compared to men, while radiotherapy is more frequently administered to men. Although surgery is the most effective treatment option in early stage disease, the lower frequency of surgical resection in men does not explain the observed survival benefit for women as a comparison of patients who received surgery as their initial treatment showed that women have better survival than men [14,31,78]. A 1995 meta-analysis reported that in advanced metastatic NSCLC, platinum-based chemotherapy provides

20 a survival benefit compared with best supportive care [79]. Platinum combinations including paclitaxel, docetaxel, vinorelbine, or gemcitabine, all offer similar outcomes for the first-line treatment of patients with advanced disease. Non-platinum combinations, such as paclitaxel or docetaxel plus gemcitabine, also result in similar survival [80—84]. Clinical factors that influence the outcome of advanced NSCLC patients treated with standard first-line chemotherapy include skin or liver metastases, loss of appetite, low performance status, four or more metastatic sites, and no prior surgery [85]. Clearly, novel treatment approaches are required to improve survival in advanced NSCLC. The role of angiogenesis is well established in lung cancer tumor progression. High microvessel density and expression of the pro-angiogenic factor vascular endothelial growth factor (VEGF) are associated with metastasis and poor survival [86,87]. Targeted inhibition of VEGF prevents the formation of new tumor vasculature and may also produce vascular normalization, allowing enhanced delivery of cytotoxic therapies to the tumor [88]. Bevacizumab is a humanized monoclonal antibody that acts by binding and neutralizing all VEGF isoforms. The efficacy and safety of bevacizumab in combination with carboplatin and paclitaxel has been assessed in patients with advanced or recurrent NSCLC (non-squamous only) in the Eastern Cooperative Oncology Group (ECOG) E4599 study. The results demonstrated a significantly increased response rate in favor of the addition of bevacizumab (10% versus 27%, P < 0.0001) and increased progression-free survival and overall survival [89]. A subset analysis of survival by sex, however, showed that the survival advantage associated with the addition of bevacizumab was male-specific (8.7 months versus 11.7 months, P = 0.001); women had similar survival in both arms, despite a significant improvement of response rate and time to progression in the bevacizumab-arm [90]. Possibly, survival may have been confounded by second- or third-line treatment as the EGFR-inhibitors are particularly active in certain patient subsets as discussed below. The small molecule inhibitors gefitinib and erlotinib prevent activation of EGFR signaling pathways by binding to the EGFR tyrosine kinase domain. As a single agent, these tyrosine kinase inhibitors induce responses in approximately 10% of relapsed patients with advanced NSCLC. In the case of gefitinib, single agent treatment in relapsed patients or combination treatment with standard chemotherapy in the first-line setting were not associated with improved survival [91—93]. Erlotinib improved survival as a single agent compared to placebo in previously treated patients, but neither erlotinib or gefitinib improved survival in combination treatment with standard chemotherapy [94,95]. While gefitinib and erlotinib fail to show activity in most patients, a dramatic and durable response is observed in a subset of patients, particularly women, Japanese, non-smokers, and patients with adenocarcinoma [96]. Correlative studies established a relationship between the presence of EGFRmutations and sensitivity to gefitinib; EGFR-mutations are present in 20% of female patients compared to only 9% of male patients [62,97]. The activity of gefitinib in selected patients is illustrative of the heterogeneous nature of lung cancer and the need to define treatment options by both clinical characteristics and tumor biology.

C.P. Belani et al. Paclitaxel poliglumex (PPX) is a macromolecular drug conjugate that links paclitaxel with a biodegradable polymer, poly-L-glutamic acid [98]. PPX is stable in systemic circulation and takes advantage of hyperpermeable tumor vasculature and suppressed lymphatic clearance to passively accumulate in tumor tissue (enhanced permeation and retention (EPR) effect). The efficacy and safety of PPX as a single agent or in combination with carboplatin was evaluated in chemotherapy-na¨ıve patients with advanced NSCLC and ECOG performance status 2 (PS2) in two phase III studies, enrolling over 800 patients [99,100]. The primary efficacy endpoint of both studies was survival. While patients receiving PPX had similar overall survival compared with the control arms, a trend towards improved survival was noted for female patients receiving PPX compared to female patients in the control arm. In contrast, survival was similar for male patients, regardless of treatment. Estrogen could potentially modulate the efficacy of PPX through the enhanced activity of cathepsin B, a ratelimiting enzyme in PPX metabolism. Alternatively, estrogen may enhance the distribution of PPX to ER-beta expressing tissues, such as lung. As a follow-up on these observations, a female-only phase III trial is underway to compare the efficacy of PPX to that of paclitaxel in women with advanced NSCLC and PS2 (PIONEER). This is the first study to take advantage of sex-based differences in lung tumor biology to optimize treatment for a subset of patients, in this case women.

6. Conclusion The reviewed epidemiological and biological lung cancer data highlight the impact of sex as a variable on various aspects of the disease: • Female sex and genetic susceptibility are among the risk factors associated with the development of lung cancer in either smokers or non-smokers. • Lung cancer tumor histology and biology vary with exposure to tobacco smoke and sex. • Variations in tumor biology may affect effectiveness of therapy; some therapies may be more effective in women than men. To further improve survival in this disease, the panel made the following recommendations: • Greater attention by the public, policy makers, and researchers on ways to reduce lung cancer risks and rates of occurrence, improve diagnosis, and expand treatment options through research—–particularly for women. • Significant increases in public and private funding to support sex- and gender-based research. • Health care provider education to reduce nihilism, pessimism, and stigma in the treatment of lung cancer patients.

Conflict of interest statement The roundtable was hosted by the Society for Women’s Health Research and supported by an unrestricted

Lung cancer in women educational grant provided by CTI Inc., Seattle. No conflicts of interest were declared by the authors.

References [1] de Braud F, Khayat D, Kroon BB, Valdagni R, Bruzzi P, Cascinelli N. Malignant melanoma. Crit Rev Oncol Hematol 2003;47:35—63. [2] Fernandez E, Bosetti C, La Vecchia C, Levi F, Fioretti F, Negri E. Sex differences in colorectal cancer mortality in Europe, 1955—1996. Eur J Cancer Prev 2000;9:99—104. [3] Stabile LP, Siegfried JM. Sex and gender differences in lung cancer. J Gend Specif Med 2003;6:37—48. [4] Jemal A, Murray T, Ward E, Samuels A, Tiwari RC, Ghafoor Ac. Cancer statistics, 2005. CA Cancer J Clin 2005;55:10—30. [5] Gandhi M, Aweeka F, Greenblatt RM, Blaschke TF. Sex differences in pharmacokinetics and pharmacodynamics. Annu Rev Pharmacol Toxicol 2004;44:499—523. [6] Anderson GD. Sex and racial differences in pharmacological response: where is the evidence? Pharmacogenetics, pharmacokinetics, and pharmacodynamics. J Womens Health (Larchmt) 2005;14:19—29. [7] Schiebinger L. Women’s health and clinical trials. J Clin Invest 2003;112:973—7. [8] Du W, Gadgeel SM, Simon MS. Predictors of enrollment in lung cancer clinical trials. Cancer 2006;106:420—5. [9] Murthy VH, Krumholz HM, Gross CP. Participation in cancer clinical trials: race-, sex-, and age-based disparities. JAMA 2004;291:2720—6. [10] Anderson RN, Arias E. The effect of revised populations on mortality statistics for the United States, 2000. Natl Vital Stat Rep 2003;51:1—24. [11] National Center for Health Statistics. Health, United States; 2005. http://www.cdc.gov/nchs/hus.htm [accessed May 28, 2006]. [12] Turgeon JL, McDonnell DP, Martin KA, Wise PM. Hormone therapy: physiological complexity belies therapeutic simplicity. Science 2004;304:1269—73. [13] Surveillance, epidemiology and end results (SEER) data base Fast Stats: lung and bronchus cancer. http://seer.cancer.gov/ [accessed May 28, 2006]. [14] Fu JB, Kau TY, Severson RK, Kalemkerian GP. Lung cancer in women: analysis of the national surveillance, epidemiology, and end results database. Chest 2005;127:768—77. [15] O’Connell JP, Kris MG, Gralla RJ, Groshen S, Trust A, Fiore JJ, et al. Frequency and prognostic importance of pretreatment clinical characteristics in patients with advanced non-smallcell lung cancer treated with combination chemotherapy. J Clin Oncol 1986;4:1604—14. [16] Ferguson MK, Skosey C, Hoffman PC, Golomb HM. Sexassociated differences in presentation and survival in patients with lung cancer. J Clin Oncol 1990;8:1402—7. [17] Shinkai T, Eguchi K, Sasaki Y, Tamura T, Ohe Y, Kojima A, et al. A prognostic-factor risk index in advanced non-smallcell lung cancer treated with cisplatin-containing combination chemotherapy. Cancer Chemother Pharmacol 1992;30: 1—6. [18] Albain KS, Crowley JJ, LeBlanc M, Livingston RB. Survival determinants in extensive-stage non-small-cell lung cancer: the Southwest Oncology Group experience. J Clin Oncol 1991;9:1618—26. [19] Paesmans M, Sculier JP, Libert P, Bureau G, Dabouis G, Thiriaux J, et al. Prognostic factors for survival in advanced nonsmall-cell lung cancer: univariate and multivariate analyses including recursive partitioning and amalgamation algorithms in 1,052 patients. The European Lung Cancer Working Party. J Clin Oncol 1995;13:1221—30.

21 [20] Johnson BE, Steinberg SM, Phelps R, Edison M, Veach SR, Ihde DC. Female patients with small cell lung cancer live longer than male patients. Am J Med 1988;85:194—6. [21] Ebbert JO, Williams BA, Sun Z, Aubry MC, Wampfler JA, Garces YI, et al. Duration of smoking abstinence as a predictor for non-small-cell lung cancer survival in women. Lung Cancer 2005;47:165—72. [22] Goodman MT, Kolonel LN, Wilkens LR, Yoshizawa CN, Le Marchand L. Smoking history and survival among lung cancer patients. Cancer Causes Control 1990;1:155—63. [23] de Perrot M, Licker M, Bouchardy C, Usel M, Robert J, Spiliopoulos A. Sex differences in presentation, management, and prognosis of patients with non-small cell lung carcinoma. J Thorac Cardiovasc Surg 2000;119:21—6. [24] Visbal AL, Williams BA, Nichols 3rd FC, Marks RS, Jett JR, Aubry MC, et al. Gender differences in non-small-cell lung cancer survival: an analysis of 4618 patients diagnosed between 1997 and 2002. Ann Thorac Surg 2004;78:209—15, discussion 215. [25] Nordquist LT, Simon GR, Cantor A, Alberts WM, Bepler G. Improved survival in never-smokers vs. current smokers with primary adenocarcinoma of the lung. Chest 2004;126: 347—51. [26] Ringer G, Smith JM, Engel AM, Hendy MP, Lang J. Influence of sex on lung cancer histology, stage, and survival in a midwestern United States tumor registry. Clin Lung Cancer 2005;7:180—2. [27] Centers for Disease Control and Prevention (CDC). http:// www.cdc.gov/tobacco/data.htm [accessed May 28, 2006]. [28] Warren CW, Jones NR, Eriksen MP, Asma S. Patterns of global tobacco use in young people and implications for future chronic disease burden in adults. Lancet 2006;367:749—53. [29] Simantov E, Schoen C, Klein JD. Health-compromising behaviors: why do adolescents smoke or drink?: identifying underlying risk and protective factors. Arch Pediatr Adolesc Med 2000;154:1025—33. [30] Saules KK, Pomerleau CS, Snedecor SM, Mehringer AM, Shadle MB, Kurth C, et al. Relationship of onset of cigarette smoking during college to alcohol use, dieting concerns, and depressed mood: results from the Young Women’s Health Survey. Addict Behav 2004;29:893—9. [31] Moore R, Doherty D, Chamberlain R, Khuri F. Sex differences in survival in non-small cell lung cancer patients 1974—1998. Acta Oncol 2004;43:57—64. [32] Chen KY, Chang CH, Yu CJ, Kuo SH, Yang PC. Distribution according to histologic type and outcome by gender and age group in Taiwanese patients with lung carcinoma. Cancer 2005;103:2566—74. [33] Muscat JE, Wynder EL. Lung cancer pathology in smokers, exsmokers and never smokers. Cancer Lett 1995;88:1—5. [34] Thun MJ, Lally CA, Flannery JT, Calle EE, Flanders WD, Heath Jr CW. Cigarette smoking and changes in the histopathology of lung cancer. J Natl Cancer Inst 1997;89:1580—6. [35] Stellman SD, Muscat JE, Thompson S, Hoffmann D, Wynder EL. Risk of squamous cell carcinoma and adenocarcinoma of the lung in relation to lifetime filter cigarette smoking. Cancer 1997;80:382—8. [36] Gorlova OY, Zhang Y, Schabath MB, Lei L, Zhang Q, Amos CI, et al. Never smokers and lung cancer risk: a case-control study of epidemiological factors. Int J Cancer 2006;118:1798—804. [37] Environmental protection agency (EPA). Respiratory health effects of passive smoking: lung cancer and other disorders; 1992. http://cfpub.epa.gov/ncea/cfm/ets/etsindex.cfm [accessed May 28, 2006]. [38] Wenzlaff AS, Cote ML, Bock CH, Land SJ, Santer SK, Schwartz DR, et al. CYP1A1 and CYP1B1 polymorphisms and risk of lung cancer among never smokers: a population-based study. Carcinogenesis 2005;26:2207—12.

22 [39] Wenzlaff AS, Cote ML, Bock CH, Land SJ, Schwartz AG. GSTM1, GSTT1 and GSTP1 polymorphisms, environmental tobacco smoke exposure and risk of lung cancer among never smokers: a population-based study. Carcinogenesis 2005;26:395—401. [40] Miller DP, De Vivo I, Neuberg D, Wain JC, Lynch TJ, Su L, et al. Association between self-reported environmental tobacco smoke exposure and lung cancer: modification by GSTP1 polymorphism. Int J Cancer 2003;104:758—63. [41] Wu AH, Fontham ET, Reynolds P, Greenberg RS, Buffler P, Liff J, et al. Family history of cancer and risk of lung cancer among lifetime nonsmoking women in the United States. Am J Epidemiol 1996;143:535—42. [42] Schwartz AG, Yang P, Swanson GM. Familial risk of lung cancer among nonsmokers and their relatives. Am J Epidemiol 1996;144:554—62. [43] Matakidou A, Eisen T, Houlston RS. Systematic review of the relationship between family history and lung cancer risk. Br J Cancer 2005;93:825—33. [44] Li X, Hemminki K. Familial multiple primary lung cancers: a population-based analysis from Sweden. Lung Cancer 2005;47:301—7. [45] Liu Y, Inoue M, Sobue T, Tsugane S. Reproductive factors, hormone use and the risk of lung cancer among middle-aged never-smoking Japanese women: a large-scale populationbased cohort study. Int J Cancer 2005;117:662—6. [46] Xu H, Spitz MR, Amos CI, Shete S. Complex segregation analysis reveals a multigene model for lung cancer. Hum Genet 2005;116:121—7. [47] Schwartz AG, Ruckdeschel JC. Familial lung cancer: genetic susceptibility and relationship to chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2006;173: 16—22. [48] International Early Lung Cancer Action Program InvestigatorsHenschke CI, Yip R, Miettinen OS. Women’s susceptibility to tobacco carcinogens and survival after diagnosis of lung cancer. JAMA 2006;296:180—4. [49] DeMarini DM. Genotoxicity of tobacco smoke and tobacco smoke condensate: a review. Mutat Res 2004;567:447—74. [50] Smith LE, Denissenko MF, Bennett WP, Li H, Amin S, Tang M, et al. Targeting of lung cancer mutational hotspots by polycyclic aromatic hydrocarbons. J Natl Cancer Inst 2000;92:803—11. [51] Mollerup S, Berge G, Baera R, Skaug V, Hewer A, Phillips DH, et al. Sex differences in risk of lung cancer: expression of genes in the PAH bioactivation pathway in relation to smoking and bulky DNA adducts. Int J Cancer 2006;119:741—51. [52] Ng DP, Tan KW, Zhao B, Seow A. CYP1A1 polymorphisms and risk of lung cancer in non-smoking Chinese women: influence of environmental tobacco smoke exposure and GSTM1/T1 genetic variation. Cancer Causes Control 2005;16:399—405. [53] Hung RJ, Boffetta P, Brockmoller J, Butkiewicz D, Cascorbi I, Clapper ML, et al. CYP1A1 and GSTM1 genetic polymorphisms and lung cancer risk in Caucasian non-smokers: a pooled analysis. Carcinogenesis 2003;24:875—82. [54] Dresler CM, Fratelli C, Babb J, Everley L, Evans AA, Clapper ML. Gender differences in genetic susceptibility for lung cancer. Lung Cancer 2000;30:153—60. [55] Husgafvel-Pursiainen K, Boffetta P, Kannio A, Nyberg F, Pershagen G, Mukeria A, et al. p53 mutations and exposure to environmental tobacco smoke in a multicenter study on lung cancer. Cancer Res 2000;60:2906—11. [56] Hainaut P, Pfeifer GP. Patterns of p53 G–>T transversions in lung cancers reflect the primary mutagenic signature of DNAdamage by tobacco smoke. Carcinogenesis 2001;22:367—74. [57] Toyooka S, Tsuda T, Gazdar AF. The TP53 gene, tobacco exposure, and lung cancer. Hum Mutat 2003;21:229—39. [58] Kure EH, Ryberg D, Hewer A, Phillips DH, Skaug V, Baera R, et al. p53 mutations in lung tumours: relationship to gender and lung DNA adduct levels. Carcinogenesis 1996;17:2201—5.

C.P. Belani et al. [59] Guinee Jr DG, Travis WD, Trivers GE, De Benedetti VM, Cawley H, Welsh JA, et al. Gender comparisons in human lung cancer: analysis of p53 mutations, anti-p53 serum antibodies and CerbB-2 expression. Carcinogenesis 1995;16:993—1002. [60] Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004;350:2129—39. [61] Hsieh RK, Lim KH, Kuo HT, Tzen CY, Huang MJ. Female sex and bronchioloalveolar pathologic subtype predict EGFR mutations in non-small cell lung cancer. Chest 2005;128: 317—21. [62] Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497—500. [63] Sugio K, Uramoto H, Ono K, Oyama T, Hanagiri T, Sugaya M, et al. Mutations within the tyrosine kinase domain of EGFR gene specifically occur in lung adenocarcinoma patients with a low exposure of tobacco smoking. Br J Cancer 2006;94: 896—903. [64] Nelson HH, Christiani DC, Mark EJ, Wiencke JK, Wain JC, Kelsey KT. Implications and prognostic value of K-ras mutation for early-stage lung cancer in women. J Natl Cancer Inst 1999;91:2032—8. [65] Soung YH, Lee JW, Kim SY, Seo SH, Park WS, Nam SW, et al. Mutational analysis of EGFR and K-RAS genes in lung adenocarcinomas. Virchows Arch 2005;446:483—8. [66] Levin ER. Integration of the extranuclear and nuclear actions of estrogen. Mol Endocrinol 2005;19:1951—9. [67] Pietras RJ, Marquez DC, Chen HW, Tsai E, Weinberg O, Fishbein M. Estrogen and growth factor receptor interactions in human breast and non-small cell lung cancer cells. Steroids 2005;70:372—81. [68] Stabile LP, Lyker JS, Gubish CT, Zhang W, Grandis JR, Siegfried JM. Combined targeting of the estrogen receptor and the epidermal growth factor receptor in non-small cell lung cancer shows enhanced antiproliferative effects. Cancer Res 2005;65:1459—70. [69] Razandi M, Pedram A, Park ST, Levin ER. Proximal events in signaling by plasma membrane estrogen receptors. J Biol Chem 2003;278:2701—12. [70] Bunone G, Briand PA, Miksicek RJ, Picard D. Activation of the unliganded estrogen receptor by EGF involves the MAP kinase pathway and direct phosphorylation. EMBO J 1996;15:2174—83. [71] Massaro D, Massaro GD. Estrogen regulates pulmonary alveolar formation, loss, and regeneration in mice. Am J Physiol Lung Cell Mol Physiol 2004;287:L1154—9. [72] Massaro D, Massaro GD. Estrogen receptor regulation of pulmonary alveolar dimensions: alveolar sexual dimorphism in mice. Am J Physiol Lung Cell Mol Physiol 2005;290:L866—70. [73] Wu CT, Chang YL, Shih JY, Lee YC. The significance of estrogen receptor beta in 301 surgically treated non-small cell lung cancers. J Thorac Cardiovasc Surg 2005;130:979—86. [74] Stabile LP, Davis AL, Gubish CT, Hopkins TM, Luketich JD, Christie N, et al. Human non-small cell lung tumors and cells derived from normal lung express both estrogen receptor alpha and beta and show biological responses to estrogen. Cancer Res 2002;62:2141—50. [75] Hershberger PA, Vasquez AC, Kanterewicz B, Land S, Siegfried JM, Nichols M. Regulation of endogenous gene expression in human non-small cell lung cancer cells by estrogen receptor ligands. Cancer Res 2005;65:1598—605. [76] Schwartz AG, Prysak GM, Murphy V, Lonardo F, Pass H, Schwartz J, et al. Nuclear estrogen receptor beta in lung cancer: expression and survival differences by sex. Clin Cancer Res 2005;11:7280—7.

Lung cancer in women [77] Kawai H, Ishii A, Washiya K, Konno T, Kon H, Yamaya C, et al. Estrogen receptor alpha and beta are prognostic factors in non-small cell lung cancer. Clin Cancer Res 2005;11:5084—9. [78] Batevik R, Grong K, Segadal L, Stangeland L. The female gender has a positive effect on survival independent of background life expectancy following surgical resection of primary non-small cell lung cancer: a study of absolute and relative survival over 15 years. Lung Cancer 2005;47:173—81. [79] Non-small Cell Lung Cancer Collaborative Group. Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials. Br Med J 1995;311:899—909. [80] Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002;346:92—8. [81] Fossella F, Pereira JR, von Pawel J, Pluzanska A, Gorbounova V, Kaukel E, et al. Randomized, multinational, phase III study of docetaxel plus platinum combinations versus vinorelbine plus cisplatin for advanced non-small-cell lung cancer: the TAX 326 study group. J Clin Oncol 2003;21:3016—24. [82] Smit EF, van Meerbeeck JP, Lianes P, Debruyne C, Legrand C, Schramel F, et al. Three-arm randomized study of two cisplatin-based regimens and paclitaxel plus gemcitabine in advanced non-small-cell lung cancer: a phase III trial of the European Organization for Research and Treatment of Cancer Lung Cancer Group–EORTC 08975. J Clin Oncol 2003;21:3909—17. [83] Georgoulias V, Papadakis E, Alexopoulos A, Tsiafaki X, Rapti A, Veslemes M, et al. Platinum-based and non-platinum-based chemotherapy in advanced non-small-cell lung cancer: a randomised multicentre trial. Lancet 2001;357:1478—84. [84] Gridelli C, Gallo C, Shepherd FA, Illiano A, Piantedosi F, Robbiati SF, et al. Gemcitabine plus vinorelbine compared with cisplatin plus vinorelbine or cisplatin plus gemcitabine for advanced non-small-cell lung cancer: a phase III trial of the Italian GEMVIN investigators and the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 2003;21:3025—34. [85] Hoang T, Xu R, Schiller JH, Bonomi P, Johnson DH. Clinical model to predict survival in chemonaive patients with advanced non-small-cell lung cancer treated with thirdgeneration chemotherapy regimens based on eastern cooperative oncology group data. J Clin Oncol 2005;23:175—83. [86] Fontanini G, Vignati S, Boldrini L, Chine S, Silvestri V, Lucchi M, et al. Vascular endothelial growth factor is associated with neovascularization and influences progression of nonsmall cell lung carcinoma. Clin Cancer Res 1997;3:861—5. [87] O’Byrne KJ, Koukourakis MI, Giatromanolaki A, Cox G, Turley H, Steward WP, et al. Vascular endothelial growth factor, platelet-derived endothelial cell growth factor and angiogenesis in non-small-cell lung cancer. Br J Cancer 2000;82:1427—32. [88] Willett CG, Boucher Y, di Tomaso E, Duda DG, Munn LL, Tong RT, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med 2004;10:145—7. [89] Sandler AB, Gray R, Brahmer J, Dowlati A, Schiller JH, Perry MC, et al. Randomized phase II/III trial of paclitaxel

23

[90]

[91]

[92]

[93]

[94]

[95]

[96]

[97]

[98] [99]

[100]

[101]

(P) plus carboplatin (C) with or without bevacizumab (NSC #704865) in patients with advanced non-squamous non-small cell lung cancer (NSCLC): an Eastern Cooperative Oncology Group (ECOG) Trial—–E4599. ASCO Annual Meeting Proceedings, vol. 23, no. 16S, Part I of II (June 1 Supplement), 2005. J Clin Oncol 2005:4. Brahmer JR, Gray R, Schiller JH, Perry M, Sandler A, Johnson D. ECOG 4599 phase III trial of carboplatin and paclitaxel ±bevacizumab: subset analysis of survival by gender. ASCO Annual Meeting Proceedings Part I, vol. 24, no. 18S (June 20 Supplement), 2006. J Clin Oncol 2006:7036. Thatcher N, Chang A, Parikh P, Rodrigues Pereira J, Ciuleanu T, von Pawel J, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced nonsmall-cell lung cancer: results from a randomised, placebocontrolled, multicentre study (Iressa survival evaluation in lung cancer). Lancet 2005;366:1527—37. Herbst RS, Giaccone G, Schiller JH, Natale RB, Miller V, Manegold C, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial—–INTACT 2. J Clin Oncol 2004;22:785—94. Giaccone G, Herbst RS, Manegold C, Scagliotti G, Rosell R, Miller V, et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial—–INTACT 1. J Clin Oncol 2004;22:777—84. Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, et al. Erlotinib in previously treated nonsmall-cell lung cancer. N Engl J Med 2005;353:123—32. Herbst RS, Prager D, Hermann R, Fehrenbacher L, Johnson BE, Sandler A, et al. TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol 2005;23:5892—9. Siegel-Lakhai WS, Beijnen JH, Schellens JH. Current knowledge and future directions of the selective epidermal growth factor receptor inhibitors erlotinib (Tarceva) and gefitinib (Iressa). Oncologist 2005;10:579—89. Bell DW, Lynch TJ, Haserlat SM, Harris PL, Okimoto RA, Brannigan BW, et al. Epidermal growth factor receptor mutations and gene amplification in non-small-cell lung cancer: molecular analysis of the IDEAL/INTACT gefitinib trials. J Clin Oncol 2005;23:8081—92. Singer JW. Paclitaxel poliglumex (XYOTAX, CT-2103): a macromolecular taxane. J Control Release 2005;109:120—6. O’Byrne K. Paclitaxel poliglumex vs. gemcitabine or vinorelbine for the treatment of performance status (PS) 2 patients with chemotherapy-na¨ıve advanced non-small cell lung cancer (NSCLC): the STELLAR 4 phase III study. Eur J Cancer 2005;3(Suppl.):324. Langer CJ, Socinski MA, O’Byrne KJ. Paclitaxel poliglumex (PPX)/carboplatin vs. paclitaxel/carboplatin for the treatment of PS2 patients with chemotherapy-na¨ıve advanced nonsmall cell lung cancer (NSCLC): a phase III study. ASCO Annual Meeting Proceedings Part I, vol. 23, no. 16S, Part I of II (June 1 Supplement), 2005. J Clin Oncol 2005:7011. Radzikowska E, Glaz P, Roszkowski K. Lung cancer in women: age, smoking, histology, performance status, stage, initial treatment and survival. Population-based study of 20 561 cases. Ann Oncol 2002;13:1087—93.