Abstract
In the field of oncology, clinical molecular diagnostics and biomarker discoveries are constantly advancing as the intricate molecular mechanisms that transform a normal cell into an aberrant state in concert with the dysregulation of alternative complementary pathways are increasingly understood. Progress in biomarker technology, coupled with the companion clinical diagnostic laboratory tests, continue to advance this field, where individualized and customized treatment appropriate for each individual patient define the standard of care. Here, we discuss the current commonly used predictive pharmacogenetic biomarkers in clinical oncology molecular testing: BRAF V600E for vemurafenib in melanoma; EML4–ALK for crizotinib and EGFR for erlotinib and gefitinib in non-small-cell lung cancer; KRAS against the use of cetuximab and panitumumab in colorectal cancer; ERBB2 (HER2/neu) for trastuzumab in breast cancer; BCR–ABL for tyrosine kinase inhibitors in chronic myeloid leukemia; and PML/RARα for all-trans-retinoic acid and arsenic trioxide treatment for acute promyelocytic leukemia.
Keywords: biomarker, cancer, clinical laboratory, clinical utility, molecular diagnostics, oncology, personalized medicine, pharmacogenetic, predictive medicine, testing
With the rapid evolution of genetic and genomic technologies revolutionizing our approach to prognosis, screening and targeting of therapies, the age of personalized and predictive medicine has not only defined how clinical practice is evolving today, but also portends to how it will be practiced in the future. Personalized medicine has its underpinning in the clinical molecular testing of biomarkers, which can be prognostic, predictive, pharmacodynamic or diagnostic [1–3]. Prognostic biomarkers have an association with clinical outcomes, such as overall survival or recurrence-free survival, independent of treatment [4–6]. Predictive biomarkers assess the likely benefit of a specific treatment to a specific patient, and thus are used to make clinical decisions [1,4,6]. Pharmacodynamic biomarkers measure the effect of a drug on the disease, whereas diagnostic biomarkers are used to establish the particular disease that is present in the patient sample [7,8]. Even though the progress in biomarker technology coupled with the companion clinical diagnostic laboratory tests will continue to advance medicine, where individualized and customized treatment appropriate for each individual patient will continue to define the standard of care, the clinical application of molecular diagnostics in predicting outcomes that may be clinically actionable has seen disproportionate acceptance and uptake across different medical fields [9]. The level of evidence for qualifying the clinical utility of any biomarker needs to be rigorous, and guidelines will undoubtedly evolve as the field advances [10].
In oncology, clinical molecular diagnostics and biomarker discoveries are constantly advancing as the intricate molecular mechanisms that transform a normal cell to an aberrant state in concert with the dysregulation of alternative complementary pathways are increasingly understood. Exploiting this knowledge of biomarkers led to the implementation of monoclonal antibodies and small-molecule tyrosine kinase inhibitors that target EGFR in colorectal cancers and non-small-cell lung carcinoma (NSCLC) [11]. Another case of the utility of predictive biomarkers comes in the anticipated use of poly-ADP (ribose) polymerase inhibitors in BRCA1/2-deficient tumors [12,13]. Even though the prostate-specific antigen test for screening in prostate cancer has limitations and is still controversial, the US FDA has approved the use of the prostate-specific antigen test along with a digital rectal exam to help detect prostate cancer and for monitoring recurrence in men aged 50 years and older. Clinicians have used cancer antigen 125 for ovarian cancer and carcinoembryonic antigen for colon cancer and other types of cancers for decades. The importance and necessity of these biomarkers are highlighted by the enormous healthcare expenditure on cancer drugs, and the estimated savings from patient selection and stratification based on the results of these biomarker diagnostic tests on predictive biomarkers with demonstrated clinical utility [14–17]. With predictive testing and patient stratification, not only is there a benefit of reducing unnecessary treatment, there is the additional benefit of avoiding toxic effects of the therapeutic regimen, thus decreasing morbidity, as in the case of trastuzumab and cardiotoxicity in breast cancer treatment [18].
The success stories of clinically useful pharmacogenetic predictive biomarkers in oncology thus far have come mostly from retrospective analyses of clinical trial data and impromptu genetic analyses, as exemplified by KRAS status and poor response to cetuximab and panitumumab [19,20]. A systematic prospective approach with current technologies available is defining how biomarker discoveries are made in tandem with drug development [21]. A variety of high-throughput approaches, including the use of massively parallel next-generation sequencing, single nucleotide polymorphism analysis and transcript profiling by microarray have been employed to discover new predictive biomarkers [22]. Even though these approaches may identify genes and proteins that correspond to disease progression or response to therapeutics, the information may be difficult to integrate with the mechanisms and pathways involved in tumor phenotype or drug action [17,23]. Thus, developing platforms that allow functional biomarkers to be rationalized in the context of mechanism and pathway for tumor killing by the drug are of utmost importance to support clinical drug development [24]. Recently, by applying a next-generation sequencing assay, the identification of novel ALK and RET gene fusions from colorectal cancer and NSCLC biopsies may eventually result in a clinically actionable predictive biomarker with further prospective clinical trials using RET kinase inhibitors [25].
Traditionally, cancer diagnosis has been classified according to anatomic origin, microscopic morphology and protein-based tests such as immunohistochemistry. Other useful means of diagnosis and monitoring include cell surface markers for leukemia and lymphoma, specific cytokine production and other nonspecific markers, such as Ig clonality in lymphoid tumors. Medical oncologists select the most appropriate therapy based on these characteristics and the extent of spread and staging of the tumor. In recent years, the clinical molecular testing of predictive pharmacogenetic biomarkers of high clinical utility has ushered in the era of personalized medicine in clinical oncology. In this review, we discuss the current commonly used predictive biomarkers in clinical molecular oncology testing (Table 1): BRAF V600E for vemurafenib in melanoma; EML4–ALK for crizotinib and EGFR for erlotinib and gefitinib in NSCLC; KRAS against the use of cetuximab and panitumumab in colorectal cancer; ERBB2 (HER2/neu) for trastuzumab in breast cancer; BCR–ABL for tyrosine kinase inhibitors in chronic myeloid leukemia (CML); and PML/RARα for all-trans-retinoic acid and arsenic trioxide treatment for acute promyelocytic leukemia (APL).
Table 1.
Clinical utility and targeted therapeutics for molecular biomarkers in oncology.
| Molecular biomarker | Prevalence | Clinical utility | Select targeted therapeutics |
|---|---|---|---|
| BRAF mutation | 40–60% metastatic melanoma; 90%: V600E mutation | Therapeutic target; prognostic | Vemurafenib GSK2118436/LGX818 RO5212054 RAF265 XL281 |
| EML4–ALK fusion gene | 5% NSCLC total; 22% of NSCLC in non- or light-smokers | Therapeutic target; prognostic | Crizotinib CH5424802 AP26113 |
| EGFR mutation | 10% NSCLC (US; 35% East Asians); more common in females and those who have never smoked vs those who have | Therapeutic target; prognostic | Erlotinib Gefitinib Afatinib CO-1686 Cetuximab |
| KRAS mutation | 15–25% lung adenocarcinoma; 40% colorectal cancer | Negative predictor of benefit to anti-EGFR therapy (antibody therapy for colorectal cancer, and small molecule inhibitor for lung cancer) | None |
| ERBB2 (HER2/neu) amplification | 20% breast cancer; 7–27% gastric cancer | Therapeutic target; prognostic | Trastuzumab Lapatinib Ertumaxomab MM-111 |
| BCR–ABL fusion gene | Detectable in 98% of chronic myelogenous leukemia and 5–20% of acute lymphoblastic leukemia | Diagnostic; therapeutic target; prognostic; minimal residual disease marker | Imatinib Dasatinib Nilotinib Radotinib INNO-406 DCC-2036 |
| PML–RARα fusion gene | >95% of acute promyelocytic leukemia | Diagnostic; therapeutic target; prognostic; minimal residual disease marker | All-trans-retinoic acid |
NSCLC: Non-small-cell lung cancer.
Clinical molecular diagnostics & cancer genetics
BRAF V600E for vemurafenib in melanoma
Melanoma is the leading cause of death from skin disease with prognosis ranging from good if detected early to poor if the cancer has spread beyond the skin and nearby lymph nodes. An understanding of the molecular pathogenesis of melanoma has provided important insights that recently led to the development of targeted therapies for specific subsets of patients with BRAF V600E mutation with metastatic melanoma. Activating mutations in BRAF are present in approximately 40–60% of advanced melanomas [26,27]. In 80–90% of cases, this activating mutation consists of the substitution of glutamic acid for valine at amino acid 600 (V600E mutation) in exon 15. Advanced melanomas with a mutation in BRAF appear to have some clinical differences that are associated with a more aggressive clinical course [27]. For this biomarker, Roche has developed an FDA-approved companion biomarker real-time PCR (RT-PCR) assay on the Roche cobas® 4800. This assay has been shown to be able to detect the mutation when the mutation constitutes only 10% of a mixture with wild-type BRAF gene (i.e., a ratio of 90:10 of wild-type:mutated BRAF). Hairy-cell leukemia is an uncommon hematological malignancy (2% of all leukemia) with good prognosis, characterized by accumulation of abnormal B lymphocytes. Whole exome sequencing has identified five mis-sense somatic clonal mutations, including a heterozygous mutation in BRAF that results in the V600E variant [28,29]. Tiacci et al. and Boyd et al. have also demonstrated sensitive, reliable, high-resolution melting analysis and allele-specific PCR qualitative assays to confirm the V600E mutation in hairy-cell leukemia [30,31].
Vemurafenib is a specific inhibitor of activated BRAF, and has been shown to significantly increase survival in melanoma patients whose tumor contains a V600E mutation in the BRAF gene [32]. Vemurafenib produces rapid tumor regression in the vast majority of patients with V600E-mutant melanoma, including those with extensive tumor burden and significant disease-related symptoms. Overall survival was significantly increased in patients assigned to vemurafenib compared with dacarbazine (estimated 6-month survival rates: 84 vs 64%; hazard ratio for death: 0.37; 95% CI: 0.26–0.55). Progression-free survival was significantly longer in those initially treated with vemurafenib (median: 5.3 vs 1.6 months; hazard ratio: 0.26; 95% CI: 0.20–0.33). The objective response rate was significantly higher with vemurafenib (48 vs 4%). Continued treatment appears necessary to maintain efficacy. Side effects are typically modest and do not generally restrict or limit treatment. Another specific BRAF inhibitor, GSK2118436, has also shown significant activity in patients with metastatic melanoma and is undergoing Phase III study [32,33]. BRAF testing and inhibition is also potentially relevant to other cancers in which BRAF mutations are common, such as papillary cancer of the thyroid.
EML4–ALK for crizotinib in NSCLC
Another major development in molecular diagnostic and clinical oncology is the discovery of the EML4–ALK fusion oncogen, which arises from an inversion on the short arm of chromosome 2, inv(2)(p21p23) that joins exons 1–13 of EML4 to exons 20–29 of ALK [34]. The resulting chimeric protein, EML4–ALK, contains an N-terminus derived from EML4 and a C-terminus containing the entire intracellular tyrosine kinase domain of ALK. This fusion oncogene rearrangement defines a distinct clinicopathologic subset of NSCLC, with overall incidence of approximately 5% [35–37]. However, in never or light smokers, the frequency of ALK positivity was approximately 22%, and among never or light smokers who did not have an EGFR mutation, the frequency was 33% [38]. Compared with small-cell carcinoma, NSCLCs are relative insensitive to chemotherapy. When possible, NSCLCs are surgically resected, although neoadjuvant and adjuvant chemotherapy are increasingly used. NSCLCs mostly tend to be squamous cell carcinoma, large cell carcinoma and adenocarcinoma, with the ALK mutation composing a small fraction of all NSCLC cases.
It only took 4 short years from the publication of the discovery of ALK-rearranged NSCLC to the conditional approval by the FDA of the EML4–ALK inhibitor, crizotinib, in August 2011. This may not have happened without the rapid development and validation of the companion diagnostic test Vysis ALK Break Apart FISH Probe Kit (CE-marked from Abbott Molecular; approved in September 2010). To identify ALK rearrangements, FISH is performed on formalin-fixed, paraffin-embedded tumors and defined as positive if >15% of tumor cells have split signals. The break-apart probes include two differently colored (red and green) probes that flank the highly conserved translocation breakpoint within ALK. In wild-type cells, the overlying red and green probes result in a yellow (fused) signal while in ALK-rearranged cells, these probes are separated and individual red and green signals are observed. Other methods to identify ALK activation include immunohistochemistry, which is not yet the gold standard [39]. The EML4–ALK fusion transcript can also be demonstrated with RT-PCR [40]. The efficacy of crizotinib is quite impressive; a recent report observed an overall response rate of 57% and rate of stable disease of 33% [41]. Historically, the response rate in NSCLC in the second-line setting is approximately 10% [42].
EGFR for erlotinib & gefitinib in NSCLC
The EGF receptor (EGFR) is a transmembrane protein with cytoplasmic kinase activity that transduces important growth factor signals from the extracellular milieu to the cell. Early nonmetastatic NSCLCs are usually not very sensitive to chemotherapy or radiation, so surgery is the preferred treatment of choice with adjuvant chemotherapy. For advanced metastatic NSCLC, a wide variety of chemotherapies are used. Studies have suggested that for advanced NSCLC patients with EGFR-mutant tumors, initial therapy with a tyrosine kinase inhibitor instead of chemotherapy may be the best choice of treatment [43]. In the USA, the currently available EGFR tyrosine kinase inhibitor, erlotinib, is FDA-approved as monotherapy for NSCLC as a second-line, and in the maintenance setting irrespective of EGFR status based on modest but statistically significant improvement in overall and progression-free survival, as demonstrated in the SATURN maintenance study (ClinicalTrials.gov identifier: NCT01194050 [101]) and NCIC BR.21 trial (ClinicalTrials.gov identifier: NCT00036647 [102]) in relapsed setting [44,45]. Therefore, mutation testing has become the standard of care to identify these patients.
DNA mutational analysis is the preferred method to assess EGFR status. Over the years, a multitude of techniques have been developed with varying sensitivities to detect known and de novo mutations, with differing instrumentations, reagents, assay runtimes and costs [46]. The peptide nucleic acid-locked nucleic acid PCR clamp is one method capable of detecting EGFR mutations in a background of wild-type EGFR [47]. This method employs fluorescent primers with preferred amplification of the mutant sequence, which is then detected by locked nucleic acids to increase specificity. The sensitivity and specificity of the peptide nucleic acid-locked nucleic acid PCR clamp method are 97 and 100%, respectively [47]. Many commercial EGFR mutation detection kits are available, such as from Genzyme and QIAGEN. The Roche cobas® EGFR Mutation Test is CE-marked, and identifies 41 mutations across exons 18, 19, 20 and 21. Currently, tumor samples are required, whether for primary lung lesion or metastasis, and are normally formalin-fixed and paraffin-embedded. One future direction is the testing of surrogate samples such as bronchial alveolar lavage, sputum, pleural fluid or serum.
KRAS against cetuximab or panitumumab in colorectal cancer
In contrast to the above biomarkers that select patients who can benefit from applications of molecular-targeted treatments, the KRAS mutation in colorectal cancer selects against patients who will not benefit from anti-EGFR receptor therapy, namely cetuximab or panitumumab. Even though colorectal cancer is one of the most common causes of cancer in both sexes, with increased and effective screening strategies prognosis is good with early detection. Mutations in the KRAS oncogene are overexpressed in colorectal cancer but are common in many other types of cancers, including pancreatic, lung, and ovarian cancer (~20%) [48]. It is a key element in the MAPK, JAK–STAT and PI3K cell-signaling pathways, acting as a key signal transducer for a number of cellular receptors. Mutations in the gene lead to abnormal cellular growth, proliferation and differentiation as a result of the activation of cell signaling. Most methods utilized to detect mutations, including nucleic acid sequencing, allele-specific PCR methods, single-strand conformational polymorphism analysis, melt-curve analysis and probe hybridization, employ PCR to amplify exons 2 and 3 of the gene, and distinguish wild-type from mutant sequences in key codons 12 and 13 [49]. No specific methodology is recommended, as all methods in current clinical use appear to have adequate clinical sensitivity for predicting a lack of response to cetuximab and panitumumab [50].
ERBB2 (HER2/neu) for trastuzumab in breast cancer
Breast cancer is responsible for approximately 14% of cancer deaths in women worldwide. The prognosis and survival rate vary greatly depending on the sex and geographical location of the patient, as well as cancer type, staging and treatment. Mutations in BRCA1 and BRCA2 account for 5–10% of breast cancers in Caucasian women in the USA [51–53]. The 185delAG and 5382insC mutations in BRCA1, and 6174delT mutation in BRCA2, are most commonly found in the Ashkenazi Jewish population [54]. Mutations in several other genes, such as TP53, PTEN, CHEK2, MLH1 and MSH2 are also associated with hereditary forms of breast cancers [55,56].
The hormone receptor status of the tumor, whether estrogen receptor (ER) or progesterone receptor, can predict the outcome to suppression therapy with tamoxifen or raloxifene [57,58]. Tamoxifen competes with estrogen for binding to the ER and has been used as first-line therapy for decades. If the tumor is hormone receptor-negative, then hEGF receptor 2 (HER2/neu) status will determine the efficacy of trastuzumab and lapatinib. There is evidence of crosstalk between ER and HER2/neu signaling pathways during breast cancer development, leading to the expression of multiple receptors [59]. Aromatase inhibitors, such as third-generation letrozole, have been shown to be more effective than tamoxifen at blocking tumor progression, independent of HER2/neu [60]. In breast cancers coexpressing HER2/neu and hormone receptor, aromatase inhibitors in combination with lapatinib have been shown to significantly improve disease outcome [60]. A serine protease inhibitor targeting the urokinase plasminogen activator system is currently in Phase II trial in patients with metastatic, HER2-negative breast cancer [61]. Before the FDA revoked its conditional approval of bevacizumab in 2011, the monoclonal antibody against VEGF-A was used in conjunction with chemotherapy for HER2-negative metastatic breast cancer [62].
Mechanisms of distinguishing breast cancer subtypes include histopathology and molecular pathology. In the last 15 years, microarrays have allowed for the study of the expression of multiple genes and the use of expression patterns as an indicator of breast cancer progression [63,64]. An emerging area of research is the correlation of biomarkers with the behavior of breast cancer subtypes. In the more aggressive triple-negative breast cancers, the presence of biomarker PKCα or the lack of nuclear biomarker ERβ, is associated with more aggressive breast cancer behavior, endocrine resistance and poorer prognosis [65]. Another recent discovery suggests that 14–3–3 theta/tau and tBID can predict neoadjuvant chemotherapy resistance in ER-positive breast cancers [66]. Upregulation of heat shock protein 90 (HSP90) mRNA expression appears to predict the aggressive behavior of HER2-negative breast cancer [67]. BRCA1-inter-ribosomal entry sequence overexpression is associated with mechanisms directed at avoiding apoptosis, and triggers aggressive tumor formation, especially in HER2-positive or triple-negative/basal-like breast cancers [68].
BCR–ABL for tyrosine kinase inhibitors in CML
CML is a clonal bone marrow stem cell disorder characterized by the proliferation of myeloid cells in the bone marrow. The understanding of the molecular pathogenesis of CML and the development of therapy to target the causative molecular defect have led to dramatic improvements in patient survival and quality of life. Patients with CML have the chromosomal abnormality t(9;22)(q34;q11.2), which results in the BCR–ABL fusion gene. The enhanced tyrosine kinase activity of BCR–ABL is responsible for activation of several signal transduction pathways. This results in the leukemic phenotype of CML cells, which encompasses deregulated proliferation, reduced adherence to the bone marrow stroma and defective apoptotic response to mutagenic stimuli [69]. In 90–95% of cases, the translocation is recognized by routine karyotyping. In the remaining cases, the chromosomal rearrangement is complex or cytogenetically cryptic, and the translocation can only be detected by FISH or RT-PCR [70].
In the absence of therapy, patients with CML eventually progress from chronic phase into a transformed phase, characterized by deteriorating hematologic parameters and worsening performance status. Conventional chemotherapy improves median survival by approximately 4 years, but does little to delay the onset of accelerated phase or blast phase [71]. The understanding of the abnormal signaling in CML cells led to the design and synthesis of small molecules that target the tyrosine kinase activity of BCR–ABL, of which imatinib was the first to be successfully used. Imatinib competes with ATP for binding to the BCR–ABL kinase domain, thus preventing phosphorylation of tyrosine residues. Interruption of this oncogenic signal is very effective for control of the disease, particularly when used in the chronic phase. However, the emergence of subclones of leukemic progenitor cells with point mutations that prevent the binding of the inhibitor to the kinase domain of BCR–ABL can lead to drug resistance. The second-generation compounds, nilotinib and dasatinib, can circumvent this form of drug failure in the case of most kinase domain mutations associated with imatinib resistance [69,72].
The most important prognostic indicator is the response to treatment at the hematologic, cytogenetic and molecular level [73,74]. Currently the complete cytogenetic response rate to imatinib is 70–90%, with a 5-year progression-free survival and overall survival between 80–95% [69]. Despite the efficacy of imatinib, it is not curative, and transcripts of BCR–ABL remain detectable by quantitative RT-PCR in most patients [75]. In some hematological cancers (e.g., acute lymphoblastic leukemia, CML and APL) as opposed to solid tumors, minimal residual disease (MRD) testing determines effectiveness of treatment, compares efficacy of different treatments, and monitors patient remission status and recurrence. Therefore, patients with CML need to be continually monitored in order to detect the level of BCR–ABL transcripts in the blood or bone marrow, as well as to detect evidence of cytogenetic remission or evolution [70,75,76]. Measurement of low-level disease or MRD using molecular tests is becoming the gold standard of measuring response to therapy, owing to its higher sensitivity compared with other routine techniques. Equipment used by different laboratories vary (Applied Biosystems® 7500 and 7900, Roche’s LightCycler®, Corbett RotorGene™ [QIAGEN], Cepheid products, Stratagene products and Biorad’s CFX™ series) with the choice more often dictated by cost and workload [77].
PML/RARα for all-trans-retinoic acid and arsenic trioxide treatment in APL
APL constitutes 5–8% of acute myeloid leukemia (AML) cases, with an abnormal accumulation of promyelocytes in the blood and bone marrow [69,78]. Prompt diagnosis is essential because of the high frequency of life-threatening disseminated intravascular coagulation. The t(15;17)(q22;q12) results in fusion of the promyelocytic gene (PML) on chromosome 15 with the retinoic acid receptor (RARα) gene on chromosome 17. The PML–RARα fusion protein mediates a block in myeloid differentiation. The blasts are highly sensitive to anthracycline-based chemotherapy, and differentiate in response to all-trans-retinoic acid and arsenic trioxide treatment [78]. All-trans-retinoic acid targets the RARα component of the fusion protein, whereas arsenic trioxide targets PML, causing maturation and apoptosis.
Three breakpoint regions are described on the PML gene at band q22 of chromosome 15 [79]. Cytogenetics, FISH, monoclonal anti-PML antibodies or RT-PCR is necessary for genetic confirmation of the aberrant PML–RARα fusion [80]; RT-PCR is the only technique that can identify the PML–RARα isoform useful for the monitoring of MRD [81,82]. Quantitative RT-PCR technology improves the predictive value of MRD monitoring. It is used to assess response to treatment and evaluate prognosis of disease, and therefore guides therapy in order to reduce the rate of relapse and to increase the rate of cure in high-risk patients [83]. Sequential RT-PCR monitoring provides the strongest predictor of relapse-free survival in APL, and provides a valid strategy to reduce rates of clinical relapse when coupled with preemptive therapy [81]. In adult patients who achieve complete remission, the prognosis is better than for any other category of AML. Once considered the most malignant human leukemia associated with the worst prognosis, APL has been transformed in the past few decades into the most frequently curable, with advances in diagnostic molecular testing, sensitive MRD monitoring by PCR, definition of relapse-risk categories and adoption of risk-adapted strategies [69,84].
Summary
BRAF V600E for vemurafenib in melanoma and hairy-cell leukemia:
Activating mutations in BRAF are present in approximately 40–60% of advanced melanomas. In 80–90% of cases, this activating mutation consists of the substitution of glutamic acid for valine at amino acid 600 in exon 15;
Vemurafenib is a specific inhibitor of activated BRAF and has been shown to significantly increase survival in patients whose tumor contains a V600E mutation in the BRAF gene. The use of vemurafenib should be limited to patients whose tumor contains this mutation;
BRAF testing and inhibition is also potentially relevant to other cancers in which BRAF mutations are common, such as papillary cancer of the thyroid.
EML4–ALK for crizotinib in NSCLC:
EML4–ALK fusion oncogene arises from an inversion on the short arm of chromosome 2, inv(2)(p21p23), that joins exons 1–13 of EML4 to exons 20–29 of ALK. The resulting chimeric protein, EML4–ALK, contains an N-terminus derived from EML4 and a C-terminus containing the entire intracellular tyrosine kinase domain of ALK;
This fusion oncogene rearrangement defines a distinct clinicopathologic subset of NSCLC with an overall incidence of approximately 5%;
Conditional approval by the FDA of the EML4–ALK inhibitor, crizotinib, 4 years after the discovery of ALK-rearranged NSCLC, may not have happened without the rapid development and validation of the companion diagnostic test.
EGFR for erlotinib and gefitinib in NSCLC:
For advanced NSCLC patients with EGFR-mutant tumors, initial therapy with a tyrosine kinase inhibitor instead of chemotherapy may be the best choice of treatment.
KRAS against cetuximab or panitumumab in colorectal cancer:
The KRAS mutation in colorectal cancer selects against patients who will not benefit from anti-EGFR receptor therapy, namely cetuximab or panitumumab.
ERBB2 (HER2/neu) for trastuzumab in breast cancer:
The hormone receptor status of the tumor, whether ER or progesterone receptor, can predict the outcome of hormone suppression therapy with tamoxifen or raloxifene;
In hormone receptor-negative tumors, HER2/neu status will probably determine the efficacy of trastuzumab and lapatinib.
BCR–ABL for tyrosine kinase inhibitors in CML:
Patients with CML have the chromosomal abnormality t(9;22) (q34;q11.2), which results in the BCR–ABL fusion gene;
Imatinib competes with ATP for binding to the BCR–ABL kinase domain, thus preventing phosphorylation of tyrosine residues. Interruption of this oncogenic signal is very effective for control of the disease, particularly when used in chronic phase;
Despite the efficacy of imatinib, it is not curative, and transcripts of BCR–ABL remain detectable by quantitative RT-PCR in most patients. Therefore, patients with CML need to be monitored continually to detect the level of BCR–ABL transcripts in the blood or bone marrow, as well as to detect evidence of cytogenetic remission or evolution;
Emergence of subclones of leukemic progenitor cells with point mutations that prevent the binding of the inhibitor to the kinase domain of BCR–ABL can lead to drug resistance. The second-generation compounds nilotinib and dasatinib can circumvent this form of drug failure.
PML/RARα for all-trans-retinoic acid and arsenic trioxide treatment in APL:
APL constitutes 5–8% of AML cases;
Prompt diagnosis is essential because of the high frequency of life-threatening disseminated intravascular coagulation;
The t(15;17)(q22;q12) results in fusion of the promyelocytic gene on chromosome 15 with the retinoic acid receptor gene on chromosome 17;
The blasts are highly sensitive to anthracycline-based chemotherapy and differentiate in response to all-trans-retinoic acid and arsenic trioxide treatment.
Expert commentary
With advances and implementation of clinical molecular diagnostics, personalized medicine in oncology has taken great strides, with predictive biomarkers guiding both therapy and monitoring of disease progression or remission. The progress in biomarker technology, coupled with companion clinical diagnostic laboratory tests, will continue to advance medicine where individualized and customized treatment appropriate for each individual patient will continue to define the standard of care. Not only are they relatively less toxic with less side effects than conventional chemotherapy, some targeted therapies are also more efficacious in tumor types where conventional chemotherapy previously provided little or no benefit. Many of the targeted therapies are orally administered, and are therefore more convenient for the patient. Unfortunately, the administration of many targeted agents results primarily in partial response or stable disease, with few complete remissions, and most targeted agents are not curative. Some of the drawbacks of such clinical molecular diagnostics include potential loss or diminished value of diagnostic or prognostic biomarkers in the setting of previously instituted therapies. Furthermore, many molecular diagnostic modalities have specimen requirements that preclude the use of size-limited or pauci-cellular specimens for multiple testing, such as needle core biopsies of lung tissue for EGFR and ALK fusion testing in NSCLC. However, careful consideration of preanalytic variables and emerging technologies for clinical molecular testing will likely abrogate such issues. The elucidation of molecular biomarkers, as well as their use in design and implementation of targeted therapies, is shifting paradigms in cancer chemotherapy, from tissue- or disease-based therapeutic regimens to molecular target-based protocols. Certainly, the availability of ever-increasing molecular data sets will hasten these advancements.
Five-year view
Discovery of new biomarkers will employ high-throughput methodologies through prospective hypothesis-driven testing. With the ‘thousand-dollar’ whole-genome sequencing within reach in the next few years, it will not be the cost of genotyping or sequencing that will deter the progress of biomarker discovery and utilization. Even though these approaches may identify genes and proteins that correspond to disease progression or response to therapeutics, it may be difficult to integrate this information with the mechanisms and pathways involved in tumor phenotype or drug action. Drug development, clinical validation and eventual implementation of these biomarkers can be supported by the rationalization of biomarkers in the context of mechanism and pathway for tumor killing and drug response.
As more targets for cellular inhibition are discovered, including abnormal targets that drive malignant cell proliferation or prolong survival, the efficacy of individualized therapy will continue to improve. Normal nonmutated targets will also be uncovered, and if blocked or stimulated, will stop malignant cells from proliferation or differentiation and apoptosis. These discoveries will facilitate and boost targeted drug therapy development, such as FMS-like receptor tyrosine kinase-3 inhibitor for AML and KRAS inhibitors. Personalizd therapy will also improve with the optimization of complete pathway blockade, such as adding lapatinib and pertuzumab to trastuzumab in the HER2/neu pathway. Another future direction is the improvement of efficacy and decreased toxicity via use of antibody–drug conjugates, such as brentuximab for Hodgkin’s lymphoma and trastuzumab–DM1 for breast cancer.
Individualized tumor pharmacogenetic analysis will continue to improve. There will be more knowledge regarding the optimal dose and administration schedule of targeted agents. There will also be more clinical guidelines pertaining to combining targeted agents with conventional therapy, or using multiple targeted agents together, concomitantly or sequentially. For these advances to transpire, scientists will need to continue to pursue the molecular and genetic abnormalities of the full spectrum of tumor types, and thus cancer patients of all types are invaluable in all phases of clinical trials.
Key issues.
Personalized medicine in clinical oncology has its underpinning in the clinical molecular testing of biomarkers, which can be diagnostic, prognostic, pharmacodynamic and/or predictive.
The importance and necessity of these predictive biomarkers are highlighted by the enormous healthcare expenditure on chemotherapy, the estimated savings, and the avoidance of toxic effects and morbidity from patient selection and stratification based on clinical molecular testing.
Current commonly used predictive biomarkers of high clinical utility in clinical oncology molecular testing include: BRAF V600E for vemurafenib in melanoma; EML4–ALK for crizotinib and EGFR for erlotinib and gefitinib in non-small-cell lung cancer; KRAS against the use of cetuximab and panitumumab in colorectal cancer; ERBB2 (HER2/neu) for trastuzumab in breast cancer; BCR–ABL for tyrosine kinase inhibitors in chronic myeloid leukemia; and PML/RARα for all-trans-retinoic acid and arsenic trioxide treatment for acute promyelocytic leukemia.
The elucidation of molecular biomarkers and their use in design and implementation of targeted therapies is shifting paradigms in cancer chemotherapy, from tissue- or disease-based therapeutic regimens to molecular target-based protocols.
Individualized tumor pharmacogenetic analysis will continue to improve and will translate into optimal dosing and scheduling of targeted agents. There will also be more guidance as to combining targeted agents with conventional therapy or using multiple targeted agents together, concomitantly or sequentially.
Footnotes
For reprint orders, please contact reprints@future-science.com
Financial & competing interests disclosure
This work was supported by NIH grant F32 HL105036 (FS Ong) and Cedars–Sinai Medical Center Clinical and Translational Science Institute Clinical Scholars Award (FS Ong). 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
Papers of special note have been highlighted as:
• of interest
•• of considerable interest
- 1.August J. Market watch: emerging companion diagnostics for cancer drugs. Nat Rev Drug Discov. 2010;9(5):351. doi: 10.1038/nrd3173. [DOI] [PubMed] [Google Scholar]
- 2.Williams GH, Stoeber K. The cell cycle and cancer. J Pathol. 2012;226(2):352–364. doi: 10.1002/path.3022. [DOI] [PubMed] [Google Scholar]
- 3.Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet. 2011;378(9805):1812–1823. doi: 10.1016/S0140-6736(11)61539-0. [DOI] [PubMed] [Google Scholar]
- 4.Buyse M, Michiels S, Sargent DJ, Grothey A, Matheson A, De Gramont A. Integrating biomarkers in clinical trials. Expert Rev Mol Diagn. 2011;11(2):171–182. doi: 10.1586/erm.10.120. [DOI] [PubMed] [Google Scholar]
- 5.Park JW, Kerbel RS, Kelloff GJ, et al. Rationale for biomarkers and surrogate end points in mechanism-driven oncology drug development. Clin Cancer Res. 2004;10(11):3885–3896. doi: 10.1158/1078-0432.CCR-03-0785. [DOI] [PubMed] [Google Scholar]
- 6•.McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. Reporting recommendations for tumor marker prognostic studies. J Clin Oncol. 2005;23(36):9067–9072. doi: 10.1200/JCO.2004.01.0454. Extensive discussion of biomarkers and reporting recommendations. [DOI] [PubMed] [Google Scholar]
- 7.Clarke PA, Workman P. Phosphatidylinositide-3-kinase inhibitors: addressing questions of isoform selectivity and pharmacodynamic/predictive biomarkers in early clinical trials. J Clin Oncol. 2012;30(3):331–333. doi: 10.1200/JCO.2011.38.7167. [DOI] [PubMed] [Google Scholar]
- 8.Sarker D, Workman P. Pharmacodynamic biomarkers for molecular cancer therapeutics. Adv Cancer Res. 2007;96:213–268. doi: 10.1016/S0065-230X(06)96008-4. [DOI] [PubMed] [Google Scholar]
- 9•.Ong FS, Deignan JL, Kuo JZ, et al. Clinical utility of pharmacogenetic biomarkers in cardiovascular therapeutics: a challenge for clinical implementation. Pharmacogenomics. 2012;13(4):465–475. doi: 10.2217/pgs.12.2. Review of pharmacogenetic biomarkers in cardiovascular therapeutics. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10•.Mcshane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. Reporting recommendations for tumor marker prognostic studies (REMARK) J Natl Cancer Inst. 2005;97(16):1180–1184. doi: 10.1093/jnci/dji237. The REMARK guidelines provide relevant information about study design preplanned hypotheses, patient and specimen characteristics, assay methods and statistical analysis methods in biomarker prognostic studies. In addition the guidelines provide helpful suggestions on how to present data and important elements to include in discussions. [DOI] [PubMed] [Google Scholar]
- 11.Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. N Engl J Med. 2008;358(11):1160–1174. doi: 10.1056/NEJMra0707704. [DOI] [PubMed] [Google Scholar]
- 12.Turner N, Tutt A, Ashworth A. Targeting the DNA repair defect of BRCA tumours. Curr Opin Pharmacol. 2005;5(4):388–393. doi: 10.1016/j.coph.2005.03.006. [DOI] [PubMed] [Google Scholar]
- 13.Ashworth A. A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. J Clin Oncol. 2008;26(22):3785–3790. doi: 10.1200/JCO.2008.16.0812. [DOI] [PubMed] [Google Scholar]
- 14.Yabroff KR, Warren JL, Brown ML. Costs of cancer care in the USA: a descriptive review. Nat Clin Pract Oncol. 2007;4(11):643–656. doi: 10.1038/ncponc0978. [DOI] [PubMed] [Google Scholar]
- 15.Yabroff KR, Freedman A, Brown ML, Ballard-Barbash R, Mcneel T, Taplin S. Trends in abnormal cancer screening results in the United States of America. J Med Screen. 2007;14(2):67–72. doi: 10.1258/096914107781261909. [DOI] [PubMed] [Google Scholar]
- 16.Yabroff KR, Davis WW, Lamont EB, et al. Patient time costs associated with cancer care. J Natl Cancer Inst. 2007;99(1):14–23. doi: 10.1093/jnci/djk001. [DOI] [PubMed] [Google Scholar]
- 17.Sawyers C. Targeted cancer therapy. Nature. 2004;432(7015):294–297. doi: 10.1038/nature03095. [DOI] [PubMed] [Google Scholar]
- 18.Suter TM, Procter M, Van Veldhuisen DJ, et al. Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trial. J Clin Oncol. 2007;25(25):3859–3865. doi: 10.1200/JCO.2006.09.1611. [DOI] [PubMed] [Google Scholar]
- 19.Lievre A, Bachet JB, Le Corre D, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006;66(8):3992–3995. doi: 10.1158/0008-5472.CAN-06-0191. [DOI] [PubMed] [Google Scholar]
- 20.Linardou H, Dahabreh IJ, Kanaloupiti D, et al. Assessment of somatic K-RAS mutations as a mechanism associated with resistance to EGFR-targeted agents: a systematic review and meta-analysis of studies in advanced non-small-cell lung cancer and metastatic colorectal cancer. Lancet Oncol. 2008;9(10):962–972. doi: 10.1016/S1470-2045(08)70206-7. [DOI] [PubMed] [Google Scholar]
- 21.Kelloff GJ, Sigman CC. Cancer biomarkers: selecting the right drug for the right patient. Nat Rev Drug Discov. 2012;11(3):201–214. doi: 10.1038/nrd3651. [DOI] [PubMed] [Google Scholar]
- 22.Schilsky RL. Personalized medicine in oncology: the future is now. Nat Rev Drug Discov. 2010;9(5):363–366. doi: 10.1038/nrd3181. [DOI] [PubMed] [Google Scholar]
- 23.Beckman RA, Clark J, Chen C. Integrating predictive biomarkers and classifiers into oncology clinical development programmes. Nat Rev Drug Discov. 2011;10(10):735–748. doi: 10.1038/nrd3550. [DOI] [PubMed] [Google Scholar]
- 24.La Thangue NB, Kerr DJ. Predictive biomarkers: a paradigm shift towards personalized cancer medicine. Nat Rev Clin Oncol. 2011;8(10):587–596. doi: 10.1038/nrclinonc.2011.121. [DOI] [PubMed] [Google Scholar]
- 25••.Lipson D, Capelletti M, Yelensky R, et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med. 2012;18(3):382–384. doi: 10.1038/nm.2673. Applying a next-generation sequencing assay, the authors identified at least one clinically relevant genomic alteration in 59% of the samples and revealed two genes fusions: C2orf44–ALK in colorectal cancer sample and KIF5B–RET in a lung adenocarcinoma. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Smalley KS, Sondak VK. Melanoma – an unlikely poster child for personalized cancer therapy. N Engl J Med. 2010;363(9):876–878. doi: 10.1056/NEJMe1005370. [DOI] [PubMed] [Google Scholar]
- 27.Long GV, Menzies AM, Nagrial AM, et al. Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol. 2011;29(10):1239–1246. doi: 10.1200/JCO.2010.32.4327. [DOI] [PubMed] [Google Scholar]
- 28.Tiacci E, Trifonov V, Schiavoni G, et al. BRAF mutations in hairy-cell leukemia. N Engl J Med. 2011;364(24):2305–2315. doi: 10.1056/NEJMoa1014209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Arcaini L, Zibellini S, Boveri E, et al. The BRAF V600E mutation in hairy cell leukemia and other mature B-cell neoplasms. Blood. 2012;119(1):188–191. doi: 10.1182/blood-2011-08-368209. [DOI] [PubMed] [Google Scholar]
- 30.Tiacci E, Schiavoni G, Forconi F, et al. Simple genetic diagnosis of hairy cell leukemia by sensitive detection of the BRAF-V600E mutation. Blood. 2012;119(1):192–195. doi: 10.1182/blood-2011-08-371179. [DOI] [PubMed] [Google Scholar]
- 31.Boyd EM, Bench AJ, Van ‘T Veer MB, et al. High resolution melting analysis for detection of BRAF exon 15 mutations in hairy cell leukaemia and other lymphoid malignancies. Br J Haematol. 2011;155(5):609–612. doi: 10.1111/j.1365-2141.2011.08868.x. [DOI] [PubMed] [Google Scholar]
- 32•.Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364(26):2507–2516. doi: 10.1056/NEJMoa1103782. Reported on improved rates of and progression-free survival in patients with previously untreated melanoma with BRAFV600E mutation on vemurafenib. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Arkenau HT, Kefford R, Long GV. Targeting BRAF for patients with melanoma. Br J Cancer. 2011;104(3):392–398. doi: 10.1038/sj.bjc.6606030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34•.Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4–ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448(7153):561–566. doi: 10.1038/nature05945. Demonstrated a subset of non-small-cell lung cancer patients who express a transforming fusion kinase that may be a diagnostic molecular marker and therapeutic target. [DOI] [PubMed] [Google Scholar]
- 35.Martelli MP, Sozzi G, Hernandez L, et al. EML4–ALK rearrangement in non-small cell lung cancer and non-tumor lung tissues. Am J Pathol. 2009;174(2):661–670. doi: 10.2353/ajpath.2009.080755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Perner S, Wagner PL, Demichelis F, et al. EML4–ALK fusion lung cancer: a rare acquired event. Neoplasia. 2008;10(3):298–302. doi: 10.1593/neo.07878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Inamura K, Takeuchi K, Togashi Y, et al. EML4–ALK fusion is linked to histological characteristics in a subset of lung cancers. J Thorac Oncol. 2008;3(1):13–17. doi: 10.1097/JTO.0b013e31815e8b60. [DOI] [PubMed] [Google Scholar]
- 38.Shaw AT, Yeap BY, Mino-Kenudson M, et al. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4–ALK. J Clin Oncol. 2009;27(26):4247–4253. doi: 10.1200/JCO.2009.22.6993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mino-Kenudson M, Chirieac LR, Law K, et al. A novel, highly sensitive antibody allows for the routine detection of ALK-rearranged lung adenocarcinomas by standard immunohistochemistry. Clin Cancer Res. 2010;16(5):1561–1571. doi: 10.1158/1078-0432.CCR-09-2845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Takeuchi K, Choi YL, Soda M, et al. Multiplex reverse transcription-PCR screening for EML4–ALK fusion transcripts. Clin Cancer Res. 2008;14(20):6618–6624. doi: 10.1158/1078-0432.CCR-08-1018. [DOI] [PubMed] [Google Scholar]
- 41.Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363(18):1693–1703. doi: 10.1056/NEJMoa1006448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gridelli C, Ardizzoni A, Ciardiello F, et al. Second-line treatment of advanced non-small cell lung cancer. J Thorac Oncol. 2008;3(4):430–440. doi: 10.1097/JTO.0b013e318168c815. [DOI] [PubMed] [Google Scholar]
- 43•.Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated. EGFR N Engl J Med. 2010;362(25):2380–2388. doi: 10.1056/NEJMoa0909530. Patients with advanced non-small-cell-lung cancer selected for by EGFR mutations show improved progression-free survival with gefitinib compared with standard chemotherapy. [DOI] [PubMed] [Google Scholar]
- 44.Cappuzzo F, Ciuleanu T, Stelmakh L, et al. Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: a multicentre, randomised, placebo-controlled Phase 3 study. Lancet Oncol. 2010;11(6):521–529. doi: 10.1016/S1470-2045(10)70112-1. [DOI] [PubMed] [Google Scholar]
- 45.Shepherd FA, Rodrigues Pereira J, Ciuleanu T, et al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med. 2005;353(2):123–132. doi: 10.1056/NEJMoa050753. [DOI] [PubMed] [Google Scholar]
- 46.Pao W, Ladanyi M. Epidermal growth factor receptor mutation testing in lung cancer: searching for the ideal method. Clin Cancer Res. 2007;13(17):4954–4955. doi: 10.1158/1078-0432.CCR-07-1387. [DOI] [PubMed] [Google Scholar]
- 47.Nagai Y, Miyazawa H, Huqun, et al. Genetic heterogeneity of the epidermal growth factor receptor in non-small cell lung cancer cell lines revealed by a rapid and sensitive detection system, the peptide nucleic acid-locked nucleic acid PCR clamp. Cancer Res. 2005;65(16):7276–7282. doi: 10.1158/0008-5472.CAN-05-0331. [DOI] [PubMed] [Google Scholar]
- 48.Wang HL, Lopategui J, Amin MB, Patterson SD. KRAS mutation testing in human cancers: the pathologist’s role in the era of personalized medicine. Adv Anat Pathol. 2010;17(1):23–32. doi: 10.1097/PAP.0b013e3181c6962f. [DOI] [PubMed] [Google Scholar]
- 49.Anderson SM. Laboratory methods for KRAS mutation analysis. Expert Rev Mol Diagn. 2011;11(6):635–642. doi: 10.1586/erm.11.42. [DOI] [PubMed] [Google Scholar]
- 50.Monzon FA, Ogino S, Hammond ME, Halling KC, Bloom KJ, Nikiforova MN. The role of KRAS mutation testing in the management of patients with metastatic colorectal cancer. Arch Pathol Lab Med. 2009;133(10):1600–1606. doi: 10.5858/133.10.1600. [DOI] [PubMed] [Google Scholar]
- 51.Lynch HT, Silva E, Snyder C, Lynch JF. Hereditary breast cancer: part I. Diagnosing hereditary breast cancer syndromes. Breast J. 2008;14(1):3–13. doi: 10.1111/j.1524-4741.2007.00515.x. [DOI] [PubMed] [Google Scholar]
- 52.Silva E, Gatalica Z, Snyder C, Vranic S, Lynch JF, Lynch HT. Hereditary breast cancer: part II. Management of hereditary breast cancer: implications of molecular genetics and pathology. Breast J. 2008;14(1):14–24. doi: 10.1111/j.1524-4741.2007.00516.x. [DOI] [PubMed] [Google Scholar]
- 53.Begg CB, Haile RW, Borg A, et al. Variation of breast cancer risk among BRCA1/2 carriers. JAMA. 2008;299(2):194–201. doi: 10.1001/jama.2007.55-a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hartge P, Struewing JP, Wacholder S, Brody LC, Tucker MA. The prevalence of common BRCA1 and BRCA2 mutations among Ashkenazi Jews. Am J Hum Genet. 1999;64(4):963–970. doi: 10.1086/302320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Walsh T, Casadei S, Coats KH, et al. Spectrum of mutations in BRCA1, BRCA2, CHEK2, and TP53 in families at high risk of breast cancer. JAMA. 2006;295(12):1379–1388. doi: 10.1001/jama.295.12.1379. [DOI] [PubMed] [Google Scholar]
- 56.Campeau PM, Foulkes WD, Tischkowitz MD. Hereditary breast cancer: new genetic developments, new therapeutic avenues. Hum Genet. 2008;124(1):31–42. doi: 10.1007/s00439-008-0529-1. [DOI] [PubMed] [Google Scholar]
- 57.Barnadas A, Estevez LG, Lluch-Hernandez A, Rodriguez-Lescure A, Rodriguez-Sanchez C, Sanchez-Rovira P. An overview of letrozole in postmenopausal women with hormone-responsive breast cancer. Adv Ther. 2011;28(12):1045–1058. doi: 10.1007/s12325-011-0075-4. [DOI] [PubMed] [Google Scholar]
- 58.Tiwary R, Yu W, Degraffenried LA, Sanders BG, Kline K. Targeting cholesterol-rich microdomains to circumvent tamoxifen-resistant breast cancer. Breast Cancer Res. 2011;13(6):R120. doi: 10.1186/bcr3063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Arpino G, Wiechmann L, Osborne CK, Schiff R. Crosstalk between the estrogen receptor and the HER tyrosine kinase receptor family: molecular mechanism and clinical implications for endocrine therapy resistance. Endocr Rev. 2008;29(2):217–233. doi: 10.1210/er.2006-0045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ellis MJ, Coop A, Singh B, et al. Letrozole inhibits tumor proliferation more effectively than tamoxifen independent of HER1/2 expression status. Cancer Res. 2003;63(19):6523–6531. [PubMed] [Google Scholar]
- 61.Schmitt M, Harbeck N, Brunner N, et al. Cancer therapy trials employing level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn. 2011;11(6):617–634. doi: 10.1586/erm.11.47. [DOI] [PubMed] [Google Scholar]
- 62.Von Minckwitz G, Eidtmann H, Rezai M, et al. Neoadjuvant chemotherapy and bevacizumab for HER2-negative breast cancer. N Engl J Med. 2012;366(4):299–309. doi: 10.1056/NEJMoa1111065. [DOI] [PubMed] [Google Scholar]
- 63.Farmer P, Bonnefoi H, Becette V, et al. Identification of molecular apocrine breast tumours by microarray analysis. Oncogene. 2005;24(29):4660–4671. doi: 10.1038/sj.onc.1208561. [DOI] [PubMed] [Google Scholar]
- 64.Hu Z, Fan C, Oh DS, et al. The molecular portraits of breast tumors are conserved across microarray platforms. BMC Genomics. 2006;7:96. doi: 10.1186/1471-2164-7-96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Tonetti DA, Gao W, Escarzaga D, Walters K, Szafran A, Coon JS. PKCalpha and ERbeta are associated with triple-negative breast cancers in African American and Caucasian patients. Int J Breast Cancer. 2012;2012:740353. doi: 10.1155/2012/740353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hodgkinson VC, ELFadl D, Agarwal V, et al. Proteomic identification of predictive biomarkers of resistance to neoadjuvant chemotherapy in luminal breast cancer: a possible role for 14–3–3 theta/tau and tBID? J Proteomics. 2012;75(4):1276–1283. doi: 10.1016/j.jprot.2011.11.005. [DOI] [PubMed] [Google Scholar]
- 67.Cheng Q, Chang JT, Geradts J, et al. Amplification and high-level expression of heat shock protein 90 marks aggressive phenotypes of human epidermal growth factor receptor 2 negative breast cancer. Breast Cancer Res. 2012;14(2):R62. doi: 10.1186/bcr3168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Shimizu Y, Luk H, Horio D, et al. BRCA1-IRES overexpression promotes formation of aggressive breast cancers. PLoS ONE. 2012;7(4):e34102. doi: 10.1371/journal.pone.0034102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Swerdlow SH. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. International Agency for Research on Cancer; Lyon, France: 2008. [Google Scholar]
- 70.Quintas-Cardama A, Cortes JE. Chronic myeloid leukemia: diagnosis and treatment. Mayo Clin Proc. 2006;81(7):973–988. doi: 10.4065/81.7.973. [DOI] [PubMed] [Google Scholar]
- 71.Geary CG. The story of chronic myeloid leukaemia. Br J Haematol. 2000;110(1):2–11. doi: 10.1046/j.1365-2141.2000.02137.x. [DOI] [PubMed] [Google Scholar]
- 72.Guilhot F, Roy L, Tomowiak C. Current treatment strategies in chronic myeloid leukemia. Curr Opin Hematol. 2012;19(2):102–109. doi: 10.1097/MOH.0b013e32834ff610. [DOI] [PubMed] [Google Scholar]
- 73.Marin D, Ibrahim AR, Lucas C, et al. Assessment of BCR–ABL1 transcript levels at 3 months is the only requirement for predicting outcome for patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors. J Clin Oncol. 2012;30(3):232–238. doi: 10.1200/JCO.2011.38.6565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Baccarani M, Saglio G, Goldman J, et al. Evolving concepts in the management of chronic myeloid leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood. 2006;108(6):1809–1820. doi: 10.1182/blood-2006-02-005686. [DOI] [PubMed] [Google Scholar]
- 75.Quintas-Cardama A, Kantarjian HM, Cortes JE. Mechanisms of primary and secondary resistance to imatinib in chronic myeloid leukemia. Cancer Control. 2009;16(2):122–131. doi: 10.1177/107327480901600204. [DOI] [PubMed] [Google Scholar]
- 76.Hughes T, Deininger M, Hochhaus A, et al. Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: review and recommendations for harmonizing current methodology for detecting BCR–ABL transcripts and kinase domain mutations and for expressing results. Blood. 2006;108(1):28–37. doi: 10.1182/blood-2006-01-0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Foroni L, Gerrard G, Nna E, et al. Technical aspects and clinical applications of measuring BCR–ABL1 transcripts number in chronic myeloid leukemia. Am J Hematol. 2009;84(8):517–522. doi: 10.1002/ajh.21457. [DOI] [PubMed] [Google Scholar]
- 78.Wang ZY, Chen Z. Acute promyelocytic leukemia: from highly fatal to highly curable. Blood. 2008;111(5):2505–2515. doi: 10.1182/blood-2007-07-102798. [DOI] [PubMed] [Google Scholar]
- 79.Lo-Coco F, Ammatuna E. The biology of acute promyelocytic leukemia and its impact on diagnosis and treatment. Hematology Am Soc Hematol Educ Program. 2006;2006:156–161. 514. doi: 10.1182/asheducation-2006.1.156. [DOI] [PubMed] [Google Scholar]
- 80.Villamor N, Costa D, Aymerich M, et al. Rapid diagnosis of acute promyelocytic leukemia by analyzing the immunocytochemical pattern of the PML protein with the monoclonal antibody PG-M3. Am J Clin Pathol. 2000;114(5):786–792. doi: 10.1309/J6PU-3XY6-R0C3-NW26. [DOI] [PubMed] [Google Scholar]
- 81•.Grimwade D, Jovanovic JV, Hills RK, et al. Prospective minimal residual disease monitoring to predict relapse of acute promyelocytic leukemia and to direct pre-emptive arsenic trioxide therapy. J Clin Oncol. 2009;27(22):3650–3658. doi: 10.1200/JCO.2008.20.1533. Prospective minimal residual disease monitoring illustrates a model of an individualized approach to management. [DOI] [PubMed] [Google Scholar]
- 82.Lewis C, Patel V, Abhyankar S, et al. Microgranular variant of acute promyelocytic leukemia with normal conventional cytogenetics, negative PML/RARA FISH and positive PML/RARA transcripts by RT-PCR. Cancer Genet. 2011;204(9):522–523. doi: 10.1016/j.cancergen.2011.09.001. [DOI] [PubMed] [Google Scholar]
- 83.Reiter A, Lengfelder E, Grimwade D. Pathogenesis, diagnosis and monitoring of residual disease in acute promyelocytic leukaemia. Acta Haematol. 2004;112(1–2):55–67. doi: 10.1159/000077560. [DOI] [PubMed] [Google Scholar]
- 84.Lo-Coco F, Cicconi L. History of acute promyelocytic leukemia: a tale of endless revolution. Mediterr J Hematol Infect Dis. 2011;3(1):e2011067. doi: 10.4084/MJHID.2011.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
Websites
- 101.ClinicalTrials.gov. An Observational Study of Tarceva (Erlotinib) in Routine Practice For First Line Maintenance Therapy in Patients With Non Small Cell Lung Cancer. http://clinicaltrials.gov/ct2/show/NCT01194050.
- 102.ClinicalTrials.gov. OSI-774 (Tarceva) in Treating Patients With Stage III or Stage IV Non-Small Cell Lung Cancer. http://clinicaltrials.gov/ct2/show/NCT00036647.
