Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease subtype that has a poor prognosis. Extensive epidemiological evidence demonstrates clear socioeconomic and demographic associations with increased likelihood of TNBC in both poorer and minority populations. Thus, biological aggressiveness with few known therapeutic directions generates disparities in breast cancer outcomes for vulnerable populations. Emerging molecular evidence of potential targets in triple-negative subpopulations offers great potential for future clinical trial directions. However, trials must appropriately consider populations at risk for aggressive subtypes of disease in order to address this disparity most completely. New US FDA draft guidance documents provide both flexible outcomes for accelerated approvals as well as flexibility in design with adaptive trials. Careful planning with design, potential patient population and choices of molecular targets informed by biomarkers will be critical to address TNBC clinical care.
Keywords: clinical trials, disparities, triple-negative breast cancer
Triple-negative breast cancer (TNBC) is diagnosed when the estrogen receptor (ER), progesterone receptor (PR) and HER-2 biomarkers are determined to be negative and, in this setting, chemotherapy has been the primary therapeutic approach, particularly in the case of positive lymph node status. Recent work has identified six molecular subtypes of TNBC with varying sensitivities to known chemotherapies. Additional preclinical work has established numerous biomarkers and potential therapeutic targets such as phosphate and tensin homolog and INPBB4, components of the PI(3)K and RAS–RAF–MEK pathways, KRAS, BRAF and EGF receptor (EGFR), FGF receptor (FGFR) 1, FGFR2, IGF receptor 1, KIT, MET and alpha-type PDGF receptor and HIF1-α/ARNT. It is clear from all of these efforts that there is no one primary target that will be able to adequately target the majority of TNBC patients due to the heterogeneity of this disease.
Epidemiological evidence has demonstrated that breast cancer subtypes are most clearly associated with race, socioeconomic status and other environmental and lifestyle exposures such as stress, diet, physical activity, alcohol consumption, residential neighborhood disadvantage and occupation. Effectively, African–American women are more likely to develop TNBC than white women. Similarly, those of lower socioeconomic classes have a higher likelihood of developing TNBC than those of higher socioeconomic standing. The challenge for the cancer research community is to determine how to utilize modifiable components of these determinants in promoting cancer prevention and control strategies, for promoting appropriate inclusiveness of populations in clinical trials, and for the potential identification of therapeutic directions.
In 2012, the US FDA released a draft guidance outlining the usage of pathological complete response (pCR) as the primary outcome for high-risk early-stage neoadjuvant breast cancer clinical trials in order to promote approvals of promising therapies based on accelerated approval regulations (part 314, Code of Federal Regulations, title 21 [H] and title 601 [E]) [201]. This guidance, along with draft guidance on the use of adaptive clinical trial designs, paves the way for the development of novel clinical trial designs for targeted therapies in TNBC. Multidisciplinary efforts will be required in order to assure that design, access to populations, in-depth knowledge of available and appropriate targets and rigorous analytical approaches can be jointly applied to assure successful trial completion.
Molecular heterogeneity of breast cancer
It has been more than a decade since the seminal articles defined unique molecular subtypes of breast cancer based on transcript profiling and the resultant clinical implications that have emerged [1–3]. Disease subclassification has been practice changing in that targeted therapies can be utilized for patients with breast cancer positive for ER, PR or HER-2 gene amplification tumors [4,5]. TNBC is diagnosed when these three markers are determined to be negative, and in this setting chemotherapy has been the primary therapeutic approach, particularly in the case of positive lymph node status [4]. Specifically, triple-negative status is accurately determined, according to the American Society of Clinical Oncology and College of American Pathologists guidelines, as those breast cancers displaying an absence of the ER and PR along with HER-2 negativity defined either by immunohistochemical tissue staining or by the ratio of HER-2 gene copy number to centromere of chromosome 17 < 2 by FISH assay [6,7]. The larger subtype within TNBC is called basal-like breast cancer (BLBC), characterized by expression patterns of CK5/6, CK14, CK17, vimentin, P-cadherin, EGFR and caveolins 1 and 2, among others [8]. Currently, there is no widely accepted assay for determining BLBC, and there is imperfect concordance of TNBC and BLBC, reflecting unresolved heterogeneity of the disease classification [1–3].
Pietenpol et al. used archived gene expression data to establish and validate subtypes within TNBC [9]. Based on a training set of 386 arrays and a validation set of 201 arrays, their analysis yielded six TNBC subtypes; two basal-like (BL1 and BL2), an immunomodulatory, a mesenchymal, a mesenchymal stem-like and a luminal androgen receptor subtype. Utilizing these expression-based subtypes they characterized TNBC cell lines and demonstrated that these TNBC subtypes exhibit differential sensitivity to select chemotherapy agents in xenograft experiments. Chiosis et al. found that a number of known receptors (EGFR, IGF1R, Her3, c-Kit and Raf-1) formed a complex in TNBC with PU-H71-bound HSP90 and also identified RAF/MEK/ERK pathway components that were previously not reported to be bound to HSP90 [10].
The Cancer Genome Atlas Network recently published an analysis of primary breast cancer based on genomic DNA copy number arrays, DNA methylation, exome sequencing, mRNA arrays, miRNA sequencing and reverse-phase protein arrays [11]. In these integrated data, a molecular similarity of BLBC and serous ovarian cancer was identified, in particular loss of TP53, RB1 and BRCA1 and amplification of MYC. These data suggest common therapeutic approaches of platinum analogs and taxanes in BLBC and serous ovarian cancer. The data also demonstrate the heterogeneity of TNBCs, with approximately 20% with germline or somatic mutation of BRCA1 or BRCA2. Some of the additionally identified targets included phosphate and tensin homolog and INPBB4, and some components of the PI(3)K and RAS-RAF-MEK pathways exhibited amplification. Some other potential identified targets include KRAS, BRAF and EGFR [12,13]. Other receptor tyroskine kinases that show potential as drug targets and that were amplified in some basal-like cancers include FGFR1, FGFR2, IGF receptor 1, KIT, MET and α-type PDGF receptor, and HIF1-α/ARNT. However, there was no highly prevalent single target identified.
A recent Phase III trial failed to demonstrate a survival benefit in TNBC of the putative PARP inhibitor iniparib [14] following a successful Phase II trial [15]. However, new evidence has emerged that iniparib does not function as a PARP inhibitor [16], so more research of PARP as a molecular target is required. In addition, critical structural identification on PARP-1 and its domain interfaces required for DNA-dependent activity presents potential targets for design of novel PARP-1 inhibitors [17].
Epidemiological evidence of disparities in breast cancer & differential molecular subtype
Breast cancer continues to be the highest incident cancer among women in the USA. It is estimated that approximately 226,870 new breast cancer cases will be diagnosed among women in the USA in 2012. In addition, almost 39,510 deaths will be attributed to the disease [18]. Numerous studies have shown that although African–Americans have a lower incidence rate of breast cancer compared with whites, they have the highest mortality rates due to breast cancer compared with other racial groups. In addition, African–Americans have benefited the least from the overall decreasing trend of breast cancer mortality in recent years that has resulted from early detection and treatment [19]. Thus, significant racial disparities in breast cancer outcome continue to exist [20]. Table 1 presents a summary of the studies revealing evidence of disparities in breast cancer and its relation to tumor size, stage, grade and receptor status. While there are underlying disparities related to other features, our focus here is on the potential biological and ultimately therapeutic targeting of these disparities.
Table 1.
Epidemiological evidence of characteristics related to differential outcomes for minority breast cancer patients.
| Study (year) | Study design | Study population | Cause of disparity | Outcome | Ref. |
|---|---|---|---|---|---|
| Ademuyiwa et al. (2011) |
Review | Not applicable | Lifestyle/environment | Grade and receptor status | [20] |
| Andaya et al. (2012) |
Registry | 184,602 cases | Lifestyle/environment | Receptor status | [22] |
| Barrett et al. (2008) |
Registry | 21,516 cases | Lifestyle/environment | Stage (distant metastasis) | [30] |
| Campbell et al. (2009) |
Registry | 25,900 cases | Lifestyle/environment | Stage | [32] |
| Cho et al. (2011) | Registry | 42,714 cases | Lifestyle/environment | Stage | [33] |
| Clarke et al. (2010) | Registry | 33,199 cases | Lifestyle/environment | Receptor status | [46] |
| Clegg et al. (2009) | Registry | 20,149 cases | Lifestyle/environment | Stage | [97] |
| Cunningham et al. (2010) |
Registry | 95,159 cases | Lifestyle/environment | Receptor status and grade | [42] |
| Dai (2010) | Registry | 10,087 cases | Lifestyle/environment | Stage | [34] |
| Desantis et al. (2010) |
Registry | 193,969 cases | Lifestyle/environment | Receptor status, grade, tumor size, lymph node status and metastasis |
[43] |
| Dolle et al. (2009) | Case–control | 897 cases, 1569 controls | Lifestyle/environment | Receptor status | [23] |
| Dunn et al. (2010) | Review | Not applicable | Lifestyle/environment | Receptor status | [47] |
| Echeverria et al. (2009) |
Registry | 4589 cases | Lifestyle/environment | Stage | [35] |
| Enewold et al. (2012) |
Registry | 240,040 cases | Lifestyle/environment | Stage | [98] |
| Gerend and Pai (2008) |
Review | Not applicable | Personal health beliefs | Stage | [99] |
| Gregg (2009) | Review | Not applicable | Personal health beliefs | Stage | [100] |
| Gullatte et al. (2010) |
Retrospective | 129 cases | Personal health beliefs | Stage | [101] |
| Haas et al. (2008) | Registry | 86,723 cases | Lifestyle/environment | Stage | [36] |
| Hahn et al (2007) | Case–control (case only) |
829 cases | Lifestyle/environment | Stage | [31] |
| Halpern et al. (2007) |
Registry | 533,715 cases | Lifestyle/environment | Stage | [102] |
| Harper et al. (2009) |
Registry | Not provided | Lifestyle/environment | Stage | [103] |
| Henry et al. (2011) | Registry | 161,619 cases | Lifestyle/environment | Stage | [28] |
| Kabat et al. (2011) | Prospective | 148,030 subjects | Lifestyle/environment Receptor | status | [48] |
| Keegan et al. (2012) |
Registry | 112,256 cases | Lifestyle/environment | Receptor status | [49] |
| Keller et al. (2011) | Registry | 1,388,186 cases | Lifestyle/environment | Stage and tumor histology | [37] |
| Kouri et al. (2010) | Registry | 403,325 cases | Lifestyle/environment | Stage | [104] |
| Krieger et al. (2008) |
Registry | 42,420 cases | Lifestyle/environment | Receptor status | [51] |
| Krieger et al. (2011) |
Registry | 291,569 cases | Lifestyle/environment | Receptor status | [50] |
| Kuo et al. (2011) | Registry | 33,838 cases | Lifestyle/environment | Stage | [29] |
| Kuzmiak et al. (2008) |
Registry | 617 cases | Lifestyle/environment | Stage and tumor size | [44] |
| Kwan et al. (2009) | Prospective (case only) |
2544 cases | Lifestyle/environment | Receptor status | [52] |
| Lew et al. (2009) | Prospective | 184,418 subjects | Lifestyle/environment | Receptor status | [53] |
| Li et al. (2010) | Prospective | 87,724 subjects | Lifestyle/environment | Receptor status and tumor histology |
[54] |
| Lund et al. (2009) | Case–control (case only) |
831 cases | Lifestyle/environment | Receptor status | [26] |
| Lund et al. (2008) | Registry | 190 cases | Lifestyle/environment | Receptor status | [55] |
| Markossian et al. (2012) |
Registry | 23,500 cases | Lifestyle/environment | Stage | [38] |
| Martinez et al. (2007) |
Registry | 25,494 cases | Lifestyle/environment | Receptor status | [57] |
| McBride et al. (2007) |
Registry | 256,174 cases | Lifestyle/environment | Tumor size and positive lymph nodes |
[27] |
| Millikan et al. (2008) |
Case–control | 1424 cases, 2022 controls |
Lifestyle/environment | Receptor status | [58] |
| Morris and Mitchell (2008) |
Review | Not applicable | Lifestyle/environment | Stage and receptor status | [25] |
| Ooi et al. (2011) | Registry | 229,594 cases | Lifestyle/environment | Stage and receptor status | [56] |
| Parise et al. (2009) | Registry | 61,309 cases | Lifestyle/environment | Receptor status | [15] |
| Parise et al. (2010) | Registry | 69,358 cases | Lifestyle/environment | Stage | [64] |
| Reyes-Ortiz et al. (2008) |
Registry | 20,818 cases | Lifestyle/environment | Stage | [39] |
| Schootman et al. (2009) |
Registry | 35,937 cases | Lifestyle/environment | Distant metastases | [105] |
| Setiawan et al. (2009) |
Prospective | 84,427 subjects | Lifestyle/environment | Receptor status | [59] |
| Stark et al. (2010) | Registry | 1664 cases | Lifestyle/environment | Receptor status | [60] |
| Stead et al. (2009) | Registry | 415 cases | Lifestyle/environment | Receptor status | [61] |
| Tarlov et al. (2009) | Registry | 4533 cases | Lifestyle/environment | Stage | [40] |
| Telli et al. (2011) | Registry | 126,577 cases | Lifestyle/environment | Receptor status | [62] |
| Trivers et al. (2009) | Case only | 476 cases | Lifestyle/environment | Receptor status | [63] |
| Virnig et al. (2009) | Registry | 273,265 cases | Lifestyle/environment | Stage | [106] |
| Vona-Davis and Rose (2009) |
Review | Not applicable | Lifestyle/environment | Stage, grade and receptor status |
[45] |
| Ward et al. (2010) | Registry | 172,834 cases | Lifestyle/environment | Stage | [107] |
| Warner and Gomez (2010) |
Registry | 124,009 cases | Lifestyle/environment | Stage | [41] |
Improvements in both early detection and screening technologies have contributed to the overall decline in breast cancer mortality rates in recent years. In addition, effective systemic therapy options have been developed for the molecular marker expression of individual tumors. However, these improvements in treatment methods will lead to better health outcomes only if the disease is detected in its early stages. Tumors that are detected at advanced stages are more likely to be associated with adverse outcomes [21].
Hormone receptor status, specifically ER and PR status, has been found to be an effective prognostic indicator for overall breast cancer survival and treatment. Hormone receptor-negative tumors are more likely to be associated with recurrence and poorer survival compared with hormone receptor-positive tumors. Hormone receptor status will also indicate the effectiveness of endocrine therapy. Hormone receptor-positive tumors have been found to respond more appropriately than hormone receptor-negative tumors to hormonal treatment. Distinct differences in hormone receptor status have been shown to vary by race. Studies have found that ER-negative tumors are more frequently found among African–American women compared with white women [22–25]. These tumors are also of higher nuclear grade [26]. Tumors that do not express ER, PR and HER-2 are known as triple negative and are significantly more aggressive compared with the other subtypes. This clinically challenging tumor subtype is diagnosed more frequently among African–American women compared with white women [23–25]. It has been reported that in the USA, African–American women are twice as likely to be diagnosed with these tumors compared with white women, and that African–American women are over-represented in breast cancer patients diagnosed under the age of 45 years. The highly aggressive nature of these tumors and the lack of effective treatments contribute to the poorest overall survival observed among African–Americans [26].
A large part of the racial disparities related to breast cancer mortality can be attributed to the more advanced stage of diagnosis often observed among African–American women compared with white women [27]. A more advanced or later stage of cancer at diagnosis results in limited treatment options and reduced likelihood of survival compared with cancers diagnosed at earlier stages of disease [28]. The survival rates for distant, regional and localized stage tumors have been approximated as follows: 23.3, 83.5 and 98.3%, respectively [29]. It has consistently been shown that breast cancers among disadvantaged populations, such as racial and ethnic minorities, are more likely to remain undetected until the cancer has reached later, more advanced stages of disease [28]. Although overall breast cancer incidence is higher for white women, African–American and Hispanic women are disadvantaged regarding stage at diagnosis and survival [30]. In the USA, African–American women are more likely than white women to be diagnosed with regional or distant breast cancer [31]. McBride et al. demonstrated that tumor size is increased in African–American patients in multivariate models controlling for age and nodal status [26]. Similarly, the presence of positive nodes is elevated in African–American patients, demonstrated in multivariate analysis controlling for age and tumor size as well as ER status and other factors. These within-stage differences may contribute to the higher mortality rate found among African–Americans compared with white women [31].
Race is considered to be a significant independent risk factor for breast cancer outcomes. These racial differences in disease are considered to be multifactorial in nature since environmental as well as genetic factors are responsible for the biology of the observed tumors [25]. Several risk factors have been hypothesized in the literature to cause these racial disparities in female breast cancer biology and include socioeconomic status, stress, diet, physical activity, alcohol consumption, characteristics of neighborhood of residence and occupation [28–30,32–41].
Cho et al. found that the concentration of and increases in immigrant populations within neighborhoods influenced diagnosis of late-stage breast cancer [33]. According to Dai, residence in areas with African–American segregation significantly increased the risk of late-stage diagnosis [34]. Keller et al. found racial disparities with regard to breast cancer stage at diagnosis throughout the USA, independent of geographic location in the country. Compared with white women, nonwhite women presented with later stage disease in all geographic areas of the country examined [37].
Several studies have indicated that differences in tumor grade due to socioeconomic status contribute to outcomes disparities [20,42–44]. Ademuyiwa et al. also reported that lower serum vitamin D concentrations may be associated with higher-grade tumors [20]. Thus, the higher-grade tumors diagnosed among African–American women may be indicative of lower vitamin D levels among these women [20].
Desantis et al. concluded that African–American women were almost twice as likely to be diagnosed with large tumors compared with white women and were more likely to be lymph node-positive [43]. In addition, it was reported that insurance status and area-level educational attainment accounted for almost 31% of the excess risk of being diagnosed with a larger tumor [43]. Kuzmiak et al. also found a significant effect of insurance status on tumor size [44]. McBride et al. found that African–American women were 24% more likely to have at least one positive lymph node when compared with white women [26].
There is overwhelming evidence that racial disparities exist in relation to hormone receptor status [15,20–23,25,27,42,43–63]. Studies examining socioeconomic status as a cause of racial disparities in hormone receptor status concluded that areas of high poverty had a greater prevalence of hormone receptor-negative tumors compared with areas of low poverty. In addition, studies examining risk of triple-negative subtypes concluded that the risk of diagnosis with this subtype is greatest among African–American women compared with other races. Parise et al. examined various breast cancer subtypes in their study and found that the subtypes with the worst overall survival were all ER-negative [15]. In addition, the majority of women diagnosed with triple-negative tumors in their study were non-Hispanic African–American or Hispanic and of the lowest socioeconomic status group [64]. It is evident that the underlying tumor biology of hormone receptor-negative breast cancers has multiple potential causes, is related to socioeconomic status and race, and represents subtypes that currently pose clinical challenges.
Several studies have concluded that the characteristics of one’s neighborhood of residence can have significant effects on racial disparities in breast cancer biology outcomes. The levels of affluence and poverty in a neighborhood are significantly associated with distant metastasis at diagnosis [30]. In addition, it was found that community change also affected the likelihood of late-stage diagnosis. Cho et al. found that women residing in neighborhoods that experienced more disadvantage over time experienced additional risk of late-stage diagnosis. Increasing concentrations of immigrant populations also contributed to this additional risk. It is possible that changing population dynamics influence the likelihood of late-stage diagnosis through increasing the burden on healthcare facilities to supply adequate healthcare to residents [33].
Racial segregation in neighborhoods has also been identified as a cause of racial disparities in breast cancer biology outcome. Segregation limits the potential for employment, education and healthcare access for communities. Thus, segregation influences disparities through resulting low socioeconomic status and lack of access to adequate healthcare resources. Socioeconomic barriers in highly segregated neighborhoods, such as lack of car ownership, can impact a woman’s ability to receive a breast cancer diagnosis when the tumor is still in its early stages. In addition, Dai reported that residence in highly segregated neighborhoods is associated with the absence of personal health promotion behaviors [34]. Thus, residential segregation is a major factor associated with late stage diagnosis among African–American women due to poor mammography access and personal beliefs regarding health promotion [34].
Socioeconomic status has been shown to be strongly associated with breast cancer biology. Desantis et al. found race, insurance status and area-level educational attainment to be strongly associated with five different prognostic factors for breast cancer: hormone receptor status, grade, tumor size, lymph node status and metastasis [43]. The associations between socioeconomic characteristics and stage at diagnosis or tumor size may be explained by inadequate access to screening and lack of timely follow-up. Those who live in areas of low socioeconomic status are less likely to receive regular screening services, resulting in advanced stage at diagnosis. African–American women are more likely than white women to be of lower socioeconomic status, uninsured and are less likely to receive timely screenings. Although many studies have focused on disparities in stage at diagnosis and tumor size, we still lack sufficient knowledge regarding the effect of socioeconomic status and racial differences in tumor grade and hormone receptor status. It has been hypothesized that these racial disparities in tumor grade and hormone receptor status may be due to lifestyle-related factors such as obesity, which is more prevalent among African–American women compared with white women [43,52,61,63,65,66].
Emerging evidence also demonstrates that oral contraceptive use, reproductive and menstrual history and breast-feeding may be associated with breast cancer subtypes [67,68]. Nulliparity is associated with decreased risk of TNBC.
Thus, there is a wealth of epidemiological evidence of the potential underlying determinants of the predilection of TNBC in vulnerable populations. The challenge is for the cancer research community to determine how to utilize modifiable components of these determinants in promoting cancer prevention and control strategies, for promoting appropriate inclusiveness of populations in clinical trials, and for the potential identification of therapeutic directions. An additional challenge comes from the changing definitions of TNBC and other subtypes over time, as knowledge and techniques improve, and the fact that historic cohorts may have misclassification of patients into subtypes, further confounding efforts to understand potential risks.
Ongoing clinical trials in TNBC
In November of 2012, a search of clinicaltrials.gov revealed 81 ongoing interventional clinical trials in TNBC. Of these, 33 trials are in Phase I or Phase I/II, 39 in Phase II, three in Phase II/III and six in Phase III trials. The Phase III trials are focused on antimetabolites such as capecitabine and gemcitabine, and anthracyclines such as epirubicin, with designs focused on either the neoadjuvant or adjuvant chemotherapy setting.
The ongoing Phase I and Phase I/II trials are primarily combination therapies, such as temsirolimus (mTOR kinase inhibitor) + neratinib (dual HER-2, EGFR, tyrosine kinase inhibitor); erlotinib (tyrosine kinase inhibitor, targets EGFR) + metformin (activates AMPK) [69]; dinaciclib (selective cyclin-dependent kinases inhibitor) [70] + epirubicin hydrochloride (anthracycline chemotherapy); Notch signaling pathway inhibitor RO4929097 + paclitaxel and carboplatin; combination of BKM120 (selective PI3K inhibitor) [71–73] and olaparib (PARP inhibitor) [74–76]; veliparib (PARP inhibitor) [77–79] + cisplatin + vinorelbine ditartrate (semisynthetic vinca alkaloid antimitotic chemotherapy) [80,81]; panobinostat (HDAC inhibitor) [82,83] + letrozole [84,85]; PLX 3397 [86] and eribulin; and cediranib (EGFR tyrosine kinase inhibitor) and olaparib [74–76]. Two ongoing early trials incorporate chemotherapy with radiation therapy, including cisplatin plus radiation therapy and concurrent adjuvant carboplatin with accelerated radiotherapy.
Recent FDA guidance in breast cancer
In May of 2012, the FDA released a draft guidance [201] outlining the usage of pCR as the primary outcome for high-risk early-stage neoadjuvant breast cancer clinical trials in order to promote approvals of promising therapies based on accelerated approval regulations (part 314, Code of Federal Regulations, title 21 [H] and title 601 [E]) [87]. The draft guidance provides insight on clinical trial design and statistical considerations and characterization of drug safety, in addition to the rationale for use of pCR as a surrogate end point. Available adjuvant therapies have prolonged breast cancer survival such that trials in that setting could take more than a decade for approval in the metastatic population. Thus, the neoadjuvant setting will have the dual advantage of breast conservation as well as well-determined outcomes based on pCR. This will benefit the subpopulations of patients with substantial risk of recurrent disease, such as those with TNBC and BLBC. A meta-analysis of 14 trials comparing pre- with post-operative chemotherapy identified 50% less risk of death in patients with pCR when compared with patients with residual tumor [88]. Since TNBC patients are chemosensitive [89–91], it is anticipated that this guidance could lead to accelerated approvals for drugs to benefit this population. According to this draft guidance, randomized add-on (combination) blinded trials will be preferred, with a uniform definition of pCR, and avoidance of postoperative systemic therapy so that continued patient follow-up can more clearly inform the utility of the surrogate end point. The guidance further delineates that an analysis of pCR, disease-free survival and/or overall survival should be prespecified and planned in the protocols for those trials seeking accelerated approval status, and should incorporate the entire intent-to-treat population. The FDA also anticipates that a large difference in pCR will be necessary in order to produce a resulting statistically significant difference in disease-free survival or overall survival, but ‘large’ is not further defined and is the subject of additional research through meta-analysis.
Discussion
While it is encouraging that a great deal of effort has provided appropriate evidence for potential clinical trials for TNBC, recent progress has also demonstrated the need for novel trial design in order to appropriately account for the heterogeneity of TNBC and BLBC and potential targeted therapies. Research efforts should focus on best approaches to adaptive, flexible trial designs in the targeted therapy setting, designs that focus on the incorporation of molecular indicators for therapy selection. The I-SPY2 trial has provided a framework to begin this important direction in the neoadjuvant setting [92]. In addition, based on the epidemiological evidence, multidisciplinary approaches to the inclusion of vulnerable populations (defined by individual and area-level characteristics) in clinical trials will be particularly critical in TNBC [93,94]. The regulatory authorities have demonstrated leadership by providing two draft guidance documents, one focused on appropriate outcomes in at-risk populations for accelerated approval [201], and the other focused on adaptive design trials [95,96]. These trials will require multidisciplinary, multi-institution and multicompany alliances in order to be successful.
Future perspective
We present evidence of the molecular heterogeneity of TNBC and BLBC, diseases that presents clinical challenges of poorer survival outcomes. Epidemiological evidence is also presented that indicates African–American patients and socioeconomically disadvantaged patients are more likely to present with TNBC. Future clinical trials in TNBC will benefit from input from multiple disciplines to inform creative design, multifactorial combination targeted therapies, access to diverse populations and strength in analytics to assure the best outcomes to address the heterogeneity of disease subclassification.
Executive summary.
Molecular heterogeneity of breast cancer
∎ Triple-negative breast cancer (TNBC) is primarily diagnosed by the absence of expression of hormone receptors (estrogen receptor, progesterone receptor) and HER-2. While there is overlap with the molecularly defined basal-like breast cancer (BLBC), TNBC exhibits diversity of expression with regard to cytokeratins, EGF receptor, caveolins and other key genes that distinguish and further establish tumor heterogeneity.
Epidemiological evidence of disparities in breast cancer & differential molecular subtype
∎ TNBC and BLBC typically occur in younger women, occur more frequently in patients with germline BRCA1 mutations, and exhibit a number of adverse pathological features including higher stage and grade, with extensive epidemiological evidence of predilection to TNBC and BLBC in African–American and socioeconomically disadvantaged populations. Despite high rates of response to chemotherapy, TNBC and BLBC patients have a higher likelihood of relapse and therefore worse prognosis.
Ongoing clinical trials in TNBC
∎ Numerous targeted and combination therapies are currently under study including those regarding PARP-1/2, EGFR and kinase inhibitors. New evidence of the structural basis for PARP-1 and the domain interfaces required for DNA-dependent activity presents potential targets for design of novel PARP-1 inhibitors.
Recent US FDA guidance in breast cancer & discussion
∎ Clinical trial designs in TNBC will require multidisciplinary teams to provide insight on design, selective biomarker–target therapy relationships and access to appropriate at-risk populations, along with analytic work-flows to ensure successful clinical trials.
Acknowledgments
Financial & competing interests disclosure The authors acknowledge financial support from a Promise Grant KG091116 (H Rui, T Hyslop, T Avery) and from an Investigator Research Grant KG110710 (T Hyslop, H Rui, Y Michael, T Avery) from Komen for the Cure.
Footnotes
The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
References
- 1.Sørlie T, Tibshirani R, Parker J, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc. Natl Acad. Sci. USA. 2003;100(14):8418–8423. doi: 10.1073/pnas.0932692100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Perou CM, Sørlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–752. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
- 3.Sørlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl Acad. Sci. USA. 2001;98(19):10869–10874. doi: 10.1073/pnas.191367098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Carlson RW, Allred DC, Anderson BO, et al. Breast cancer. J. Natl Compr. Canc. Netw. 2009;7(2):122–192. doi: 10.6004/jnccn.2009.0012. [DOI] [PubMed] [Google Scholar]
- 5.Cardoso F, Bedard PL, Winer EP, et al. International guidelines for management of metastatic breast cancer: combination vs sequential single-agent chemotherapy. J. Natl Cancer Inst. 2009;101(17):1174–1181. doi: 10.1093/jnci/djp235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hammond MEH, Hayes DF, Dowsett M, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer (unabridged version) Arch. Pathol. Lab. Med. 2010;134(7):48–72. doi: 10.5858/134.7.e48. [DOI] [PubMed] [Google Scholar]
- 7.Wolff AC, Hammond MEH, Schwartz JN, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch. Pathol. Lab. Med. 2007;131(1):18–43. doi: 10.5858/2007-131-18-ASOCCO. [DOI] [PubMed] [Google Scholar]
- 8.Gluz O, Liedtke C, Gottschalk N, Pusztai L, Nitz U, Harbeck N. Triple-negative breast cancer: current status and future directions. Ann. Oncol. 2009;20(12):1913–1927. doi: 10.1093/annonc/mdp492. [DOI] [PubMed] [Google Scholar]
- 9.Lehmann BD, Bauer JA, Chen X, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Invest. 2011;121(7):2750–2765. doi: 10.1172/JCI45014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Caldas-Lopes E, Cerchietti L, Ahn JH, et al. Hsp90 inhibitor PU-H71, a multimodal inhibitor of malignancy, induces complete responses in triple-negative breast cancer models. Proc. Natl Acad. Sci. USA. 2009;106(20):8368–8373. doi: 10.1073/pnas.0903392106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Cancer Genome Atlas Network Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70. doi: 10.1038/nature11412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Corkery B, Crown J, Clynes M, O’Donovan N. Epidermal growth factor receptor as a potential therapeutic target in triple-negative breast cancer. Ann. Oncol. 2009;20(5):862–867. doi: 10.1093/annonc/mdn710. [DOI] [PubMed] [Google Scholar]
- 13.Burness ML, Grushko TA, Olopade OI. Epidermal growth factor receptor in triple-negative and basal-like breast cancer: promising clinical target or only a marker? Cancer J. 2010;16(1):23–32. doi: 10.1097/PPO.0b013e3181d24fc1. [DOI] [PubMed] [Google Scholar]
- 14.O’Shaughnessy J, Osborne C, Pippen JE, et al. Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N. Engl. J. Med. 2011;364(3):205–214. doi: 10.1056/NEJMoa1011418. [DOI] [PubMed] [Google Scholar]
- 15.Parise CA, Bauer KR, Brown MM, Caggiano V. Breast cancer subtypes as defined by the estrogen receptor (ER), progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) among women with invasive breast cancer in California, 1999–2004. Breast J. 2009;15(6):593–602. doi: 10.1111/j.1524-4741.2009.00822.x. [DOI] [PubMed] [Google Scholar]
- 16.Liu X, Shi Y, Maag DX, et al. Iniparib nonselectively modifies cysteine-containing proteins in tumor cells and is not a bona fide PARP inhibitor. Clin. Can. Res. 2012;18(2):510–523. doi: 10.1158/1078-0432.CCR-11-1973. [DOI] [PubMed] [Google Scholar]
- 17.Langelier MF, Planck JL, Roy S, Pascal JM. Structural basis for DNA damage-dependent poly (ADP-ribosyl) ation by human PARP-1. Sci. Signal. 2012;336(6082):728–732. doi: 10.1126/science.1216338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J. Clin. 2012;62(1):10–29. doi: 10.3322/caac.20138. [DOI] [PubMed] [Google Scholar]
- 19.Berz JPB, Johnston K, Backus B, et al. The influence of black race on treatment and mortality for early-stage breast cancer. Med. Care. 2009;47(9):986–992. doi: 10.1097/MLR.0b013e31819e1f2b. [DOI] [PubMed] [Google Scholar]
- 20.Ademuyiwa FO, Edge SB, Erwin DO, Orom H, Ambrosone CB, Underwood W. Breast cancer racial disparities: unanswered questions. Cancer Res. 2011;71(3):640–644. doi: 10.1158/0008-5472.CAN-10-3021. [DOI] [PubMed] [Google Scholar]
- 21.Amirikia KC, Mills P, Bush J, Newman LA. Higher population-based incidence rates of triple-negative breast cancer among young African–American women. Cancer. 2011;117(12):2747–2753. doi: 10.1002/cncr.25862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Andaya AA, Enewold L, Horner MJ, Jatoi I, Shriver CD, Zhu K. Socioeconomic disparities and breast cancer hormone receptor status. Cancer Causes Control. 2012;23(6):951–958. doi: 10.1007/s10552-012-9966-1. [DOI] [PubMed] [Google Scholar]
- 23.Dolle JM, Daling JR, White E, et al. Risk factors for triple-negative breast cancer in women under the age of 45 years. Cancer Epidemiol. Biomarkers Prev. 2009;18(4):1157–1166. doi: 10.1158/1055-9965.EPI-08-1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Morris GJ, Naidu S, Topham AK, et al. Differences in breast carcinoma characteristics in newly diagnosed African–American and Caucasian patients. Cancer. 2007;110(4):876–884. doi: 10.1002/cncr.22836. [DOI] [PubMed] [Google Scholar]
- 25.Morris GJ, Mitchell EP. Higher incidence of aggressive breast cancers in African–American women: a review. J. Natl Med. Assoc. 2008;100(6):698–702. doi: 10.1016/s0027-9684(15)31344-4. [DOI] [PubMed] [Google Scholar]
- 26.Lund M, Trivers K, Porter P, et al. Race and triple negative threats to breast cancer survival: a population-based study in Atlanta, GA. Breast Cancer Res. Treat. 2009;113(2):357–370. doi: 10.1007/s10549-008-9926-3. [DOI] [PubMed] [Google Scholar]
- 27.McBride R, Hershman D, Tsai W-Y, Jacobson JS, Grann V, Neugut AI. Within-stage racial differences in tumor size and number of positive lymph nodes in women with breast cancer. Cancer. 2007;110(6):1201–1208. doi: 10.1002/cncr.22884. [DOI] [PubMed] [Google Scholar]
- 28.Henry K, Boscoe F, Johnson C, Goldberg D, Sherman R, Cockburn M. Breast cancer stage at diagnosis: is travel time important? J. Comm. Health. 2011;36(6):933–942. doi: 10.1007/s10900-011-9392-4. [DOI] [PubMed] [Google Scholar]
- 29.Kuo T-M, Mobley LR, Anselin L. Geographic disparities in late-stage breast cancer diagnosis in California. Health Place. 2011;17(1):327–334. doi: 10.1016/j.healthplace.2010.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Barrett RE, Cho YI, Weaver KE, et al. Neighborhood change and distant metastasis at diagnosis of breast cancer. Ann. Epidemiol. 2008;18(1):43–47. doi: 10.1016/j.annepidem.2007.07.001. [DOI] [PubMed] [Google Scholar]
- 31.Hahn KME, Bondy ML, Selvan M, et al. Factors associated with advanced disease stage at diagnosis in a population-based study of patients with newly diagnosed breast cancer. Am. J. Epidemiol. 2007;166(9):1035–1044. doi: 10.1093/aje/kwm177. [DOI] [PubMed] [Google Scholar]
- 32.Campbell RT, Li X, Dolecek TA, Barrett RE, Weaver KE, Warnecke RB. Economic, racial and ethnic disparities in breast cancer in the US: towards a more comprehensive model. Health Place. 2009;15(3):870–879. doi: 10.1016/j.healthplace.2009.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cho Y, Johnson T, Barrett R, Campbell R, Dolecek T, Warnecke R. Neighborhood changes in concentrated immigration and late stage breast cancer diagnosis. J. Immigr. Minor. Health. 2011;13(1):9–14. doi: 10.1007/s10903-010-9339-3. [DOI] [PubMed] [Google Scholar]
- 34.Dai D. Black residential segregation, disparities in spatial access to health care facilities, and late-stage breast cancer diagnosis in metropolitan Detroit. Health Place. 2010;16(5):1038–1052. doi: 10.1016/j.healthplace.2010.06.012. [DOI] [PubMed] [Google Scholar]
- 35.Echeverría SE, Borrell LN, Brown D, Rhoads G. A local area analysis of Racial, ethnic, and neighborhood disparities in breast cancer staging. Cancer Epidemiol. Biomarkers Prevent. 2009;18(11):3024–3029. doi: 10.1158/1055-9965.EPI-09-0390. [DOI] [PubMed] [Google Scholar]
- 36.Haas JS, Earle CC, Orav JE, Brawarsky P, Neville BA, Williams DR. Racial segregation and disparities in cancer stage for seniors. J. Gen. Intern. Med. 2008;23(5):699–705. doi: 10.1007/s11606-008-0545-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Keller D, Guilfoyle C, Sariego J. Geographical influence on racial disparity in breast cancer presentation in the united states. Am. Surg. 2011;77(7):933–936. [PubMed] [Google Scholar]
- 38.Markossian TW, Hines RB. Disparities in late stage diagnosis, treatment, and breast cancer-related death by race, age, and rural residence among women in georgia. Women Health. 2012;52(4):317–335. doi: 10.1080/03630242.2012.674091. [DOI] [PubMed] [Google Scholar]
- 39.Reyes-Ortiz CA, Eschbach K, Zhang DD, Goodwin JS. Neighborhood composition and cancer among Hispanics: tumor stage and size at time of diagnosis. Cancer Epidemiol. Biomarkers Prevent. 2008;17(11):2931–2936. doi: 10.1158/1055-9965.EPI-07-0430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Tarlov E, Zenk S, Campbell R, Warnecke R, Block R. Characteristics of mammography facility locations and stage of breast cancer at diagnosis in Chicago. J. Urb. Health. 2009;86(2):196–213. doi: 10.1007/s11524-008-9320-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Warner E, Gomez S. Impact of neighborhood racial composition and metropolitan residential segregation on disparities in breast cancer stage at diagnosis and survival between black and white women in California. J. Comm. Health. 2010;35(4):398–408. doi: 10.1007/s10900-010-9265-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Cunningham J, Montero A, Garrett-Mayer E, Berkel H, Ely B. Racial differences in the incidence of breast cancer subtypes defined by combined histologic grade and hormone receptor status. Cancer Causes Control. 2010;21(3):399–409. doi: 10.1007/s10552-009-9472-2. [DOI] [PubMed] [Google Scholar]
- 43.Desantis C, Jemal A, Ward E. Disparities in breast cancer prognostic factors by race, insurance status, and education. Cancer Causes Control. 2010;21(9):1445–1450. doi: 10.1007/s10552-010-9572-z. [DOI] [PubMed] [Google Scholar]
- 44.Kuzmiak CM, Haberle S, Padungchaichote W, Zeng D, Cole E, Pisano ED. Insurance status and the severity of breast cancer at the time of diagnosis. Acad. Radiol. 2008;15(10):1255–1258. doi: 10.1016/j.acra.2008.04.011. [DOI] [PubMed] [Google Scholar]
- 45.Vona-Davis L, Rose DP. The influence of socioeconomic disparities on breast cancer tumor biology and prognosis: a review. J. Womens Health. 2009;18(6):883–893. doi: 10.1089/jwh.2008.1127. [DOI] [PubMed] [Google Scholar]
- 46.Clarke CA, Fish K, Kurian AW, Shema SJ. Lifetime risks of specific breast cancer subtypes among women in four racial/ethnic groups. Breast Cancer Res. (Online Edition) 2010;12:R99. doi: 10.1186/bcr2780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dunn BK, Agurs-Collins T, Browne D, Lubet R, Johnson KA. Health disparities in breast cancer: biology meets socioeconomic status. Breast Cancer Res. Treat. 2010;121(2):281–292. doi: 10.1007/s10549-010-0827-x. [DOI] [PubMed] [Google Scholar]
- 48.Kabat G, Kim M, Phipps A, et al. Smoking and alcohol consumption in relation to risk of triple-negative breast cancer in a cohort of postmenopausal women. Cancer Causes Control. 2011;22(5):775–783. doi: 10.1007/s10552-011-9750-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Keegan TH, Derouen MC, Press DJ, Kurian AW, Clarke CA. Occurrence of breast cancer subtypes in adolescent and young adult women. Breast Cancer Res. 2012;14(2):R55. doi: 10.1186/bcr3156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Krieger N, Chen JT, Waterman PD. Temporal trends in the black/white breast cancer case ratio for estrogen receptor status: disparities are historically contingent, not innate. Cancer Causes Control. 2011;22(3):511–514. doi: 10.1007/s10552-010-9710-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Krieger N, Chen JT, Ware JH, Kaddour A. Race/ethnicity and breast cancer estrogen receptor status: impact of class, missing data, and modeling assumptions. Cancer Causes Control. 2008;19(10):1305–1318. doi: 10.1007/s10552-008-9202-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kwan ML, Kushi LH, Weltzien E, et al. Epidemiology of breast cancer subtypes in two prospective cohort studies of breast cancer survivors. Breast Cancer Res. 2009;11(3):R31. doi: 10.1186/bcr2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lew JQ, Freedman ND, Leitzmann MF, et al. Alcohol and risk of breast cancer by histologic type and hormone receptor status in postmenopausal women: the NIH-AARP diet and health study. Am. J. Epidemiol. 2009;170(3):308–317. doi: 10.1093/aje/kwp120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Li CI, Chlebowski RT, Freiberg M, et al. Alcohol consumption and risk of postmenopausal breast cancer by subtype: the women’s health initiative observational study. J. Natl Cancer Inst. 2010;102(18):1422–1431. doi: 10.1093/jnci/djq316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lund MJB, Butler EN, Bumpers HL, et al. High prevalence of triple-negative tumors in an urban cancer center. Cancer. 2008;113(3):608–615. doi: 10.1002/cncr.23569. [DOI] [PubMed] [Google Scholar]
- 56.Ooi SL, Martinez ME, Li CI. Disparities in breast cancer characteristics and outcomes by race/ethnicity. Breast Cancer Res. Treat. 2011;127(3):729–738. doi: 10.1007/s10549-010-1191-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Martínez ME, Nielson CM, Nagle R, Lopez AM, Kim C, Thompson P. Breast cancer among Hispanic and non-Hispanic white women in Arizona. J. Health Care Poor Underserved. 2007;18(4):130–135. 137–145. doi: 10.1353/hpu.2007.0112. [DOI] [PubMed] [Google Scholar]
- 58.Millikan RC, Newman B, Tse CK, et al. Epidemiology of basal-like breast cancer. Breast Cancer Res. Treat. 2008;109(1):123–139. doi: 10.1007/s10549-007-9632-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Setiawan VW, Monroe KR, Wilkens LR, Kolonel LN, Pike MC, Henderson BE. Breast Cancer Risk Factors Defined by Estrogen and Progesterone Receptor Status. Am. J. Epidemiol. 2009;169(10):1251–1259. doi: 10.1093/aje/kwp036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Stark A, Kleer CG, Martin I, et al. African ancestry and higher prevalence of triple-negative breast cancer. Cancer. 2010;116(21):4926–4932. doi: 10.1002/cncr.25276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Stead LA, Lash TL, Sobieraj JE, et al. Triple-negative breast cancers are increased in black women regardless of age or body mass index. Breast Cancer Res. 2009;11(2):R18. doi: 10.1186/bcr2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Telli ML, Chang ET, Kurian AW, et al. Asian ethnicity and breast cancer subtypes: a study from the California cancer registry. Breast Cancer Res. Treat. 2011;127(2):471–478. doi: 10.1007/s10549-010-1173-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Trivers KF, Lund MJ, Porter PL, et al. The epidemiology of triple-negative breast cancer, including race. Cancer Causes Control. 2009;20(7):1071–1082. doi: 10.1007/s10552-009-9331-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Parise CA, Bauer KR, Caggiano V. Variation in breast cancer subtypes with age and race/ethnicity. Crit.Rev. Oncol./Hematol. 2010;76(1):44–52. doi: 10.1016/j.critrevonc.2009.09.002. [DOI] [PubMed] [Google Scholar]
- 65.Lin N, Vanderplas A, Hughes M, et al. Clinicopathological features and sites of recurrence according to breast cancer subtype in the National Comprehensive Cancer Network (NCCN) J. Clin. Oncol. 2009;27(Suppl. 15):543. Abstract. [Google Scholar]
- 66.Maiti B, Kundranda M, Spiro T, Daw H. The association of metabolic syndrome with triple-negative breast cancer. Breast Cancer Res. Treat. 2010;121(2):479–483. doi: 10.1007/s10549-009-0591-y. [DOI] [PubMed] [Google Scholar]
- 67.Phipps AI, Chlebowski RT, Prentice R, et al. Reproductive history and oral contraceptive use in relation to risk of triple-negative breast cancer. J. Natl Cancer Inst. 2011;103(6):470–477. doi: 10.1093/jnci/djr030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Phipps AI, Malone KE, Porter PL, Daling JR, Li CI. Reproductive and hormonal risk factors for postmenopausal luminal, HER2 overexpressing, and triple-negative breast cancer. Cancer. 2008;113(7):1521–1526. doi: 10.1002/cncr.23786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zakikhani M, Dowling R, Fantus IG, Sonenberg N, Pollak M. Metformin is an AMP kinase–dependent growth inhibitor for breast cancer cells. Cancer Res. 2006;66(21):10269–10273. doi: 10.1158/0008-5472.CAN-06-1500. [DOI] [PubMed] [Google Scholar]
- 70.Parry D, Guzi T, Shanahan F, et al. Dinaciclib (SCH 727965), a novel and potent cyclin-dependent kinase inhibitor. Mol. Cancer Ther. 2010;9(8):2344–2353. doi: 10.1158/1535-7163.MCT-10-0324. [DOI] [PubMed] [Google Scholar]
- 71.Baselga J, De Jonge M, Rodon J, et al. A first-in-human Phase I study of BKM120, an oral pan-class I PI3K inhibitor, in patients (pts) with advanced solid tumors. J. Clin. Oncol. 2010;28(Suppl. 15) doi: 10.1200/JCO.2011.36.1360. Abstract 3003. [DOI] [PubMed] [Google Scholar]
- 72.Maira SM, Pecchi S, Huang A, et al. Identification and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor. Mol. Cancer Ther. 2012;11(2):317–328. doi: 10.1158/1535-7163.MCT-11-0474. [DOI] [PubMed] [Google Scholar]
- 73.Bendell JC, Rodon J, Burris HA, et al. Phase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors. J. Clin. Oncol. 2012;30(3):282–290. doi: 10.1200/JCO.2011.36.1360. [DOI] [PubMed] [Google Scholar]
- 74.Tutt A, Robson M, Garber JE, et al. Oral poly (ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet. 2010;376(9737):235–244. doi: 10.1016/S0140-6736(10)60892-6. [DOI] [PubMed] [Google Scholar]
- 75.Tutt A, Robson M, Garber J, et al. Phase II trial of the oral PARP inhibitor olaparib in BRCA-deficient advanced breast cancer. J. Clin. Oncol. 2009;27(Suppl. 18) Abstract CRA501. [Google Scholar]
- 76.Dent R, Lindeman G, Clemons M, et al. Safety and efficacy of the oral PARP inhibitor olaparib (AZD2281) in combination with paclitaxel for the first-or second-line treatment of patients with metastatic triple-negative breast cancer: results from the safety cohort of a Phase I/II multicenter trial. J. Clin. Oncol. 2010;28(Suppl. 15) Abstract 1018. [Google Scholar]
- 77.Tan A, Toppmeyer D, Stein M, et al. Phase I trial of veliparib, (ABT-888), a poly (ADP-ribose) polymerase (PARP) inhibitor, in combination with doxorubicin and cyclophosphamide in breast cancer and other solid tumors. J. Clin. Oncol. 2011;29(Suppl.) Abstract 3041. [Google Scholar]
- 78.Lorusso P, Ji J, Li J, et al. Phase I study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of the poly (ADP-ribose) polymerase (PARP) inhibitor veliparib (ABT-888; V) in combination with irinotecan (CPT-11; Ir) in patients (pts) with advanced solid tumors. J. Clin. Oncol. 2011;29(Suppl. 15) doi: 10.1158/1078-0432.CCR-15-0652. Abstract 3000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Li X, Delzer J, Voorman R, De Morais SM, Lao Y. Disposition and drug–drug interaction potential of veliparib (ABT-888), a novel and potent inhibitor of poly (ADP-ribose) polymerase. Drug Metab. Dispos. 2011;39(7):1161–1169. doi: 10.1124/dmd.110.037820. [DOI] [PubMed] [Google Scholar]
- 80.Tsuruo T, Inaba M, Tashiro T, et al. Evaluation of antitumor activity of navelbine (vinorelbine ditartrate) against human breast carcinoma xenografts based on its pharmacokinetics in nude mice. Anticancer Drugs. 1994;5:634–640. doi: 10.1097/00001813-199412000-00004. [DOI] [PubMed] [Google Scholar]
- 81.Galano G, Caputo M, Felice Tecce M, Capasso A. Efficacy and tolerability of vinorelbine in the cancer therapy. Curr. Drug Safe. 2011;6(3):185–193. doi: 10.2174/157488611797579302. [DOI] [PubMed] [Google Scholar]
- 82.Atadja P. Development of the pan-DAC inhibitor panobinostat (LBH589): successes and challenges. Cancer Lett. 2009;280(2):233–241. doi: 10.1016/j.canlet.2009.02.019. [DOI] [PubMed] [Google Scholar]
- 83.Conte P, Campone M, Pronzato P, et al. Phase I trial of panobinostat (LBH589) in combination with trastuzumab in pretreated HER2-positive metastatic breast cancer (mBC): preliminary safety and tolerability results. J. Clin. Oncol. 2009;27(Suppl. 15) Abstract 1081S. [Google Scholar]
- 84.Ellis MJ, Coop A, Singh B, et al. Letrozole is more effective neoadjuvant endocrine therapy than tamoxifen for ErbB-1–and/or ErbB-2-positive, estrogen receptor-positive primary breast cancer: evidence from a Phase III randomized trial. J. Clin. Oncol. 2001;19(18):3808–3816. doi: 10.1200/JCO.2001.19.18.3808. [DOI] [PubMed] [Google Scholar]
- 85.Dombernowsky P, Smith I, Falkson G, et al. Letrozole, a new oral aromatase inhibitor for advanced breast cancer: double-blind randomized trial showing a dose effect and improved efficacy and tolerability compared with megestrol acetate. J. Clin. Oncol. 1998;16(2):453–461. doi: 10.1200/JCO.1998.16.2.453. [DOI] [PubMed] [Google Scholar]
- 86.Anthony S, Puzanov I, Lin P, Nolop K, West B, Von Hoff D. Pharmacodynamic activity demonstrated in Phase I for PLX3397, a selective inhibitor of FMS and Kit. J. Clin. Oncol. 2011;29(Suppl.) Abstract 3093. [Google Scholar]
- 87.Von Minckwitz G, Untch M, Blohmer JU, et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J. Clin. Oncol. 2012;30(15):1796–1804. doi: 10.1200/JCO.2011.38.8595. [DOI] [PubMed] [Google Scholar]
- 88.Van Der Hage J, Van De Velde C, Mieog S. Preoperative chemotherapy for women with operable breast cancer. Cochrane Database Syst. Rev. 2007;(2) doi: 10.1002/14651858.CD005002.pub2. CD005002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Esserman LJ, Berry DA, Cheang MCU, et al. Chemotherapy response and recurrence-free survival in neoadjuvant breast cancer depends on biomarker profiles: results from the I-SPY 1 trial (CALGB 150007/150012; ACRIN 6657) Breast Cancer Res. Treat. 2012;132(3):1–14. doi: 10.1007/s10549-011-1895-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Symmans WF, Peintinger F, Hatzis C, et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J. Clin. Oncol. 2007;25(28):4414–4422. doi: 10.1200/JCO.2007.10.6823. [DOI] [PubMed] [Google Scholar]
- 91.Liedtke C, Mazouni C, Hess KR, et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol. 2008;26(8):1275–1281. doi: 10.1200/JCO.2007.14.4147. [DOI] [PubMed] [Google Scholar]
- 92.Barker A, Sigman C, Kelloff G, Hylton N, Berry D, Esserman L. I-SPY 2: an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clin. Pharmacol. Ther. 2009;86(1):97–100. doi: 10.1038/clpt.2009.68. [DOI] [PubMed] [Google Scholar]
- 93.Advani AS, Atkeson B, Brown CL, et al. Barriers to the participation of African–American patients with cancer in clinical trials. Cancer. 2003;97(6):1499–1506. doi: 10.1002/cncr.11213. [DOI] [PubMed] [Google Scholar]
- 94.Murthy VH, Krumholz HM, Gross CP. Participation in cancer clinical trials. JAMA. 2004;291(22):2720–2726. doi: 10.1001/jama.291.22.2720. [DOI] [PubMed] [Google Scholar]
- 95.US Department of Health and Human Services . Guidance For Industry. Center for Biologics Evaluation and Research, Food and Drug Administration; MD, USA: 2010. Adaptive design clinical trials for drugs and biologics. [Google Scholar]
- 96.Hung H, O’Neill RT, Wang SJ, Lawrence J. A regulatory view on adaptive/flexible clinical trial design. Biometric. J. 2006;48(4):565–573. doi: 10.1002/bimj.200610229. [DOI] [PubMed] [Google Scholar]
- 97.Clegg L, Reichman M, Miller B, et al. Impact of socioeconomic status on cancer incidence and stage at diagnosis: selected findings from the surveillance, epidemiology, and end results: national longitudinal mortality study. Cancer Causes Control. 2009;20(4):417–435. doi: 10.1007/s10552-008-9256-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Enewold L, Zhou J, McGlynn KA, et al. Racial variation in tumor stage at diagnosis among department of defense beneficiaries. Cancer. 2012;118(5):1397–1403. doi: 10.1002/cncr.26208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gerend MA, Pai M. Social determinants of black-white disparities in breast cancer mortality: a review. Cancer Epidemiol. Biomarkers Prev. 2008;17(11):2913–2923. doi: 10.1158/1055-9965.EPI-07-0633. [DOI] [PubMed] [Google Scholar]
- 100.Gregg G. Psychosocial issues facing African and African–American women diagnosed with breast cancer. Soc. Work Public Health. 2009;24(1–2):100–116. doi: 10.1080/19371910802569609. [DOI] [PubMed] [Google Scholar]
- 101.Gullatte MM, Brawley O, Kinney A, Powe B, Mooney K. Religiosity, spirituality, and cancer fatalism beliefs on delay in breast cancer diagnosis in African–American women. J. Religion Health. 2010;49(1):62–72. doi: 10.1007/s10943-008-9232-8. [DOI] [PubMed] [Google Scholar]
- 102.Halpern MT, Bian J, Ward EM, Schrag NM, Chen AY. Insurance status and stage of cancer at diagnosis among women with breast cancer. Cancer. 2007;110(2):403–411. doi: 10.1002/cncr.22786. [DOI] [PubMed] [Google Scholar]
- 103.Harper S, Lynch J, Meersman SC, Breen N, Davis WW, Reichman MC. Trends in area–socioeconomic and race–ethnic disparities in breast cancer incidence, stage at diagnosis, screening, mortality, and survival among women ages 50 years and over. Cancer Epidemiol. Biomarkers Prev. 2009;18(1):121–131. doi: 10.1158/1055-9965.EPI-08-0679. [DOI] [PubMed] [Google Scholar]
- 104.Kouri E, He Y, Winer E, Keating N. Influence of birthplace on breast cancer diagnosis and treatment for Hispanic women. Breast Cancer Res. Treat. 2010;121(3):743–751. doi: 10.1007/s10549-009-0643-3. [DOI] [PubMed] [Google Scholar]
- 105.Schootman M, Jeffe DB, Gillanders WE, Aft R. Racial disparities in the development of breast cancer metastases among older women. Cancer. 2009;115(4):731–740. doi: 10.1002/cncr.24087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Virnig BA, Baxter NN, Habermann EB, Feldman RD, Bradley CJ. A matter of race: early- versus late-stage cancer diagnosis. Health Affairs. 2009;28(1):160–168. doi: 10.1377/hlthaff.28.1.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ward EMP, Fedewa SaMPH, Cokkinides V, Virgo K. The association of insurance and stage at diagnosis among patients aged 55–74 years in the national cancer database. Cancer J. 2010;16:614–621. doi: 10.1097/PPO.0b013e3181ff2aec. [DOI] [PubMed] [Google Scholar]
Website
- 201.Prowell T. Pathologic complete response in neoadjuvant treatment of high-risk early-stage breast cancer: use as an endpoint to support accelerated approval. Draft guidance for industry. 2012 www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/ Guidances/UCM305501.pdf.
