Next Article in Journal
Weekends-Off Lenvatinib for Unresectable Hepatocellular Carcinoma Improves Therapeutic Response and Tolerability Toward Adverse Events
Next Article in Special Issue
Current Strategies and Future Perspectives for Precision Medicine in Pancreatic Cancer
Previous Article in Journal
Public Adverse Event Data Insights into the Safety of Pembrolizumab in Melanoma Patients
Previous Article in Special Issue
Personalized Medicine—Current and Emerging Predictive and Prognostic Biomarkers in Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Personalized Medicine: Recent Progress in Cancer Therapy

by
Valentina Gambardella
1,2,
Noelia Tarazona
1,2,
Juan Miguel Cejalvo
1,
Pasquale Lombardi
3,
Marisol Huerta
1,
Susana Roselló
1,2,
Tania Fleitas
1,2,
Desamparados Roda
1,2,*,† and
Andres Cervantes
1,2,*,†
1
Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, 46010 Valencia, Spain
2
Instituto de Salud Carlos III, CIBERONC, 28220 Madrid, Spain
3
Department of Oncology, University of Turin; Candiolo Cancer Institute - FPO- IRCCS, 10060 Candiolo (TO), Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this article.
Cancers 2020, 12(4), 1009; https://doi.org/10.3390/cancers12041009
Submission received: 12 March 2020 / Revised: 5 April 2020 / Accepted: 15 April 2020 / Published: 19 April 2020
(This article belongs to the Special Issue Personalized Medicine: Recent Progress in Cancer Therapy)

Abstract

:
Translational research has revolutionized how we develop new treatments for cancer patients. The change from an organ-centric concept guiding treatment choice towards deep molecular analysis, driving a personalized approach, is one of the most important advances of modern oncology. Several tools such as next generation sequencing and RNA sequencing have greatly improved the capacity to detect predictive and prognostic molecular alterations. Detection of gene mutations, amplifications, and fusions has therefore altered the history of several diseases in both a localized and metastatic setting. This shift in perspective, in which attention is focused on the specific molecular alterations of the tumor, has opened the door to personalized treatment. This situation is reflected in the increasing number of basket trials selecting specific molecular targets. Nonetheless, some weaknesses need to be addressed. The complexity of cancer cells enriched with concomitant molecular alterations complicates identification of the driver. Moreover, tumor heterogeneity could be responsible for the lack of benefit when targeted agents are used. In light of this, there is growing interest in the role of multidisciplinary committees or molecular tumor boards to try to enhance selection. The aim of this review is to critically analyze the evolution of cancer treatment towards a precision approach, underlining some recent successes and unexpected failures.

1. The Revolution of Tumor Treatment: From Tumor Site to Molecular Alterations

During the last few years, cancer patient treatment has been completely revolutionized as several molecular alterations have been identified as drivers of cancer development and progression [1]. This biomarker-based approach has historically proceeded from basic science to validation in the clinic. Thus, to guide treatment choice, attention has gradually turned to biomarkers rather than the anatomic site of origin of a given tumor [2]. Due to this change, large groups of patients whose tumors bear a particular molecular feature have been successfully treated with a single targeted drug. The first example of molecular-based medicine for cancer patients was the use of endocrine therapy in luminal breast cancer [3]. However, the point probably marking the revolution towards a precision medicine approach was the approval of imatinib in patients with chronic myeloid leukemia bearing the t (9,22) translocation that creates a BCR-ABL fusion kinase [4].
The identification of druggable molecular alterations marked a new era in oncology [5]. The most important results were achieved in breast [6,7,8,9] and lung cancer [10,11,12] and in melanoma [13]. In breast cancer, apart from identifying hormonal receptors as therapeutic agents, other pinpointed biomarkers such as PIK3CA (Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform) or ERBB2 (human epidermal growth factor receptor 2) mutations have completely altered the therapeutic approach in luminal breast cancer patients [7,14,15,16]. Moreover, the detection of HER2 amplification as a driver has also contributed immensely to locating another important subgroup of patients who definitely benefit from anti-HER2 inhibition in all clinical settings [17,18,19]. A fundamental shift was also observed in patients diagnosed with non-small cell lung cancer (NSCLC). The identification of EGFR (Epidermal Growth Factor Receptor) mutations [20] and EML4-ALK (echinoderm microtubule associated protein-like 4- Anaplastic lymphoma kinase) translocation [21] has affected outcomes for advanced diseases. Moreover, identification of the BRAF-V600E mutation and the subsequent treatment with BRAF and MEK inhibitors [14] has significantly improved clinical outcomes in patients diagnosed with advanced melanoma. These represent only a few examples of the application of precision medicine in daily clinical practice. Indeed, the genomic revolution has impacted significantly on the majority of solid tumors nowadays, involved also in “orphan diseases” like extrahepatic cholangiocarcinoma [22].
Pinpointing specific molecular characteristics (such as microsatellite instability across all solid tumors), somatic or germline mutations, (such as BRCA, BRAF or ERBB2) [23,24,25,26], or gene fusions (FGFR and NTRK) [27,28] could guide treatment choice in this field, improving patient outcomes independently of tumor location. To properly apply these molecular-based treatments, it is ultimately necessary to distinguish which patient groups will probably benefit or not from this type of therapy by identifying specific biomarkers predictive of response or resistance [29].
Although genomics seems to be a really relevant start point to plan a precision approach for cancer patients, it is clear that molecular phenotype measurements and characterization are a needed arm to the understanding of tumor to improve the precision medicine approach. In some cases, the use of proteomic could also help when several molecular alterations are detectable, making it difficult to identify the most relevant driver to be targeted [30]. Moreover, genetic mutations do not always result in the predicted change of the corresponding protein, and there are many other factors that contribute to tumor behavior, such as protein modifications, metabolism, and the microbiome [31]. Another important field to try to improve a precision medicine approach is metabolomic [32]. The importance of it is not only for the identification of targetable biomarkers but also for the identification of pharmacological phenotype able to understand the mechanisms of inter-patient variability in response to drug therapy [33]. Metabolomics can also help in evaluating drug resistance and disease relapse [34,35], leading to opportunities for the development of novel therapeutic strategies [32].
Identifying those molecular features has led to a clinical strategy in which testing upfront or after first-line failure may provide certain novel therapeutic opportunities. In umbrella trials, patients diagnosed with the same type of solid tumors are treated according to their molecular features, while in basket trials, patients diagnosed with different types of solid tumors with a common driver molecular alteration are selected and treated with a specific inhibitor [36]. As an example, a multisite study (including >1000 patients with non-small cell lung cancer) showed that matching was associated with longer survival than was seen in patients without genotype-directed treatment. Likewise, two meta-analyses [37,38] in 70,000 patients reported that trials with a personalized strategy led to a higher proportion of responding patients and longer progression-free and overall survival than trials with unselected patients. Standout results were obtained in patients diagnosed with tumors harboring ERBB2 mutations or NTRK or FGFR fusions [15,27,28] (Figure 1).
In the MOSCATO 01 trial, cancer treatment was planned according to genomic analyses. A potentially actionable molecular alteration was identified in 411 of 843 patients. A total of 199 patients were treated with a targeted agent matched to a genomic alteration. The Progression-Free survival 2/1 (PFS2/1) ratio was >1.3 in about one-third of the patients. Although objective responses were observed in only 11% of patients, progression-free survival with this molecularly guided strategy was longer than obtained by the previous line of therapy in one-third of 194 patients. This study suggested that high-throughput genomics could improve outcomes in molecularly selected patients when treated accordingly [39].

2. Precision Molecular Oncology: Understanding the Role of New Drivers with Novel Drugs

Several potential targets for novel drugs have been identified using high-throughput technologies, and several compounds have recently been approved or are under investigation.
One of the most prevalent molecular alterations in solid tumors are PIK3CA mutations. The PI3K/AKT/mTOR (The phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR)) pathway is an intracellular signaling pathway implicated in cell proliferation. This pathway can be activated at several points, but PIK3CA mutations and PTEN (Phosphatase and tensin homolog) function loss are the most frequent detectable molecular alterations [40]. For this reason, several basket trials have been conducted or are still ongoing to assess the role of PIK3CA inhibitors in several solid tumors. In luminal breast cancer, incidence of PIK3CA mutations is about 30% in primary tumors and metastases. In a phase III trial enrolling PIK3CA mutant luminal breast cancer patients progressing to hormonal treatment, adding alpelisib, a specific p110α PIK3CA inhibitor, to fulvestrant significantly improved antitumor activity (response rate 26.6% vs. 12.8%) and progression-free survival (11 vs. 5.7 months) over a combination of placebo plus fulvestrant, leading to its regulatory approval. These results confirm that PIK3CA mutations could be drivers in metastatic luminal breast cancer and suggest the importance of detecting this molecular alteration in selected patients [7]. Other more specific novel PI3K inhibitors are currently under clinical development (Table 1).
Activation of fibroblast growth factor receptor (FGFR) pathways also plays a major role. FGFRs are made up of four highly conserved transmembrane tyrosine kinases receptors (FGFR1–4) and FGFR5, which lacks an intracellular kinase domain. All these receptors are activated by fibroblast growth factor (FGF), participating in cell survival and proliferation. Deregulation of the FGF signaling axis is implicated in oncogenesis, tumor progression, and resistance to anticancer therapy across several solid tumors, with a prevalence below 10%. Multiple trials studying diverse solid tumors have proposed the aberrant FGFR signaling pathway as a potential therapeutic target, and several inhibitors are under development. FGFR alterations may consist of amplification, mutations, and gene-fusion, yet the specific role of each one and their contribution in predicting drug response is not clear [56]. In early clinical trials, promising results were reported in patients who show FGFR2 amplification in gastric cancer and in patients with FGFR2 and FGFR3 translocations in cholangiocarcinoma and urothelial cancers, respectively. Disappointingly, only modest levels of clinical activity have been reported for patients with other aberrations, such as FGFR1 amplification7 or FGFR2 mutation, in advanced-stage endometrial cancer [57]. Urothelial cancer is one of the tumors that most harbors FGFR alterations. In particular, the luminal I subtype is characterized by a lack of benefit from immunotherapy and a higher rate of mutations in the gene encoding for FGFR, detectable in about 20% of all urothelial cancers [58]. In a phase II study [59] treating patients diagnosed with locally advanced and unresectable or metastatic urothelial carcinoma with FGFR alterations, the potent FGFR1–4 tyrosine inhibitor erdafatinib demonstrated antitumor activity, achieving objective responses in 40% of previously treated patients. Interestingly, patients who had undergone previous immunotherapy also achieved the same benefit. FGFR2 activation by point mutations and FGFR1–3 amplification or overexpression are also observed in subsets of patients with advanced cholangiocarcinoma [49]. The most common alterations are chromosomal fusions consisting of FGFR2 exons 1 to 17, encoding the intact extracellular and kinase domains, fused in-frame to a 3′ partner that possesses a protein dimerization domain. Multiple FGFR-selective inhibitors are being tested in clinical trials in cholangiocarcinoma patients harboring FGFR pathway alterations (Table 1).
BRAF mutations are major oncogenic drivers in 7% of solid tumors [60]. BRAF inhibition has led to significant improvement in melanoma. In colorectal cancer, BRAF mutation has been associated with specific clinicopathological features as well as very poor prognosis. BRAF mutations are more prevalent in proximal colon tumors, poorer differentiation, mucinous histology, and older females [61]. However, several clinical trials testing BRAF inhibitors as single agents in BRAF-mutant colon cancer patients saw response in less than 5%, mostly of short duration. This lack of efficacy was attributed to feedback EGFR activation resulting in MAPK (mitogen-activated protein kinase) signaling pathway reactivation. Based on this hypothesis, the combination of BRAF/MEK inhibitors plus monoclonal antibodies against EGFR has been explored. In a phase III trial, 665 metastatic colorectal patients with BRAF-V600E mutation who had experienced treatment failure with one or two prior regimens were recruited to receive binimetinib, encorafenib, and cetuximab versus irinotecan-based chemotherapy. This triplet therapy was well tolerated with a manageable safety profile. Moreover, it showed a better response rate than chemotherapy (26% vs. 2%) as well as significantly improved overall survival (OS) for those receiving the triple targeted agent combination (9 vs. 5.4 months), becoming a new standard of care for previously treated BRAF V600E–mutant metastatic colorectal cancer patients and underlining the need to uncover BRAF mutations in all advanced colon cancer patients [62].
Recently, NTRK fusions have also been detected as a possible target [63]. The neurotrophic tyrosine receptor kinases (NTRK) plays a key role in nervous system development. These are three receptors, TrkA, TrkB, and TrkC, with a tyrosine kinase intracellular domain able to activate signaling pathways like MAPK, PI3K/AKT, and PLCγ/PKC (phospholipase-Cγ and protein kinase C signal pathways) resulting in enhanced migration, cell differentiation, synapse formation, and proliferation. In NTRK fusions, the 3′-region is fused with a 5′ region of a fusion partner, resulting in a chimeric receptor protein with dimerization, and thus, uncontrolled activation of the TRK kinase domain leads to chimeric proteins with constitutively activated or overexpressed kinase function conferring dominant oncogenic potential. These oncogenic fusions can be either intra or interchromosomal. All three NTRK genes are potentially affected. These alterations are considered rare, being present in about 0–25%, with the highest occurrence in secretory breast cancer, mammary analog secretory cancer (MASC) of salivary glands, congenital mesoblastic nephroma and infantile fibrosarcoma thyroid cancers, spitzoid melanomas, and gastrointestinal stroma tumors (GIST) [63]. The presence of NTRK fusion seems to indicate early alterations in solid tumors, so the use of specific inhibitors would supposedly improve clinical outcomes. Larotrectinib, a selective TRK inhibitor, was tested in adults and children whose tumors harbored a NTRK 1-2-3 fusion. The overall response rate was 75% [28]. Other NTRK inhibitors are currently under investigation, and some have shown clinical activity in patients resistant to first-generation inhibitors due to the development of secondary mutations (Table 1 and Figure 2).

3. Limitations of Molecular Driven Treatments in the Clinic

Despite a promising biological background justifying precision medicine, certain negative results have highlighted limitations in the molecularly driven treatment strategy. The SHIVA trial [64] could serve as a paradigm for what is still incompletely understood when applying for precision medicine in cancer patients. In this trial, 741 patients diagnosed with any kind of solid tumor were screened for molecular alterations and randomized to receive the physician in charge’s treatment of choice versus targeted therapy selected according to their molecular profile. No benefit in median PFS was observed in molecularly oriented patients versus conventional approach (Hazard-ratio (HR) 0.88, p = 0.41), suggesting that off-label use of molecularly targeted agents does not improve progression-free survival compared with standard treatment [64]. Nevertheless, critical analysis can reveal certain limitations potentially responsible for the failure of this approach [65]. It is well known that the presence of multiple molecular alterations together with tumor heterogeneity consistently limits the use of monotherapy, which could justify the lack of benefit derived from a precision approach [66]. Moreover, elements of the approach used in this trial are questionable, such as hormonal therapy in heavily treated patients or nonspecific inhibitors when targeting certain molecular alterations [64].
In the NCI-MATCH trial, patients diagnosed with solid tumors are randomized according to their molecular alterations in 40 different arms. In three cohorts from the NCI-MATCH trial, patients with tumors harboring ERRB2 amplification, FGFR alterations, or PIK3CA mutations were treated with T-DM1, FGFR inhibitors, or taselisib, respectively. Unfortunately, response rates were very low, under 10% across several cohorts [67]. In this case, the presence of heavily pretreated populations and concomitant molecular alterations could justify the lack of benefit.
Moreover, several discordant results have been obtained using selected targets in solid tumors sharing the same molecular alterations. It is widely known for instance that blocking HER2 in breast cancer has been associated with significant improvement in clinical outcomes [3,8,19], although the use of antiHER2 agents in gastric or cancer did not confer the same results [68]. The same negative results were observed when BRAF inhibitors were used as single agent in melanoma or colorectal cancer CRC, suggesting that, aside from the molecular alterations, in these mutations, several other molecular characteristics need to be considered when applying precision medicine to cancer patients. For these reasons, tumor heterogeneity, molecular mechanisms of resistance in pretreated patients conferring a complex pattern of mutations, could cause resistance to single targeted agents and could become the final cause of the imprecision of precision medicine. Moreover, Several limitations could also derive from pharmacodynamic or kinetic aspects that could cause inter- and intraindividual variability, limiting the use of targeted agents such as gender, weight, ethnicity, and renal and hepatic functions [69]. A precision approach could be difficultly adopted when a patient presents more than one factor which could interfere with drug exposure or response. [70]. Therefore, an implementation of precision medicine should be researched. The preliminary results from the NCI-MATCH trial highlight a critical biological reality that has been known for some time: genomic alterations do not always lead to oncogenic pathway activation or addiction, and targeting multiple drivers and/or resistance pathways may be required for optimal antitumor efficacy [5,71,72]. A relevant limitation of targeted therapies is the presence of molecular alterations responsible for primary or acquired resistance [68]. This phenomenon has been widely studied and described in all solid tumors. The amplification of transmembrane receptors or intracellular proteins hyperactivation among the same pathway to which the targeted agent is driven or among other cellular pathways implicated in cancer cell proliferation and survival could be responsible for the lack of response to a specific inhibitor [73].

Limitations of the Molecular Approach

Another point that should be addressed is the evolution of the specific molecular tools used for the selection of patients enrolled in the clinical trials. In some precision medicine-based trials, patients were screened by immunohistochemistry (IHC) or PCR to evaluate the presence of a protein expression or hotspots mutation. However, nowadays, the use of a complete Next Generation Sequencing (NGS) panel able to evaluate a wide screen of molecular alterations is quite common. The analysis performed by NGS or RNA sequencing provides high sensitivity in the detection of specific molecular alterations and also the capability of detecting concomitant alterations that could cause eventually resistance to specific targeted agents [64,74] (Table 2).
A better understanding of cancer biology is needed to optimize and define context-dependent oncogenic mutations and resistance mechanisms. In this scenario, preclinical models, such as patient-derived xenografts and organoids, may help elucidate potential codrivers and resistance mechanisms so that rational combinations can be designed and tested to support clinical deployment [81]. Analyses of circulating tumor DNA may also provide insights regarding dynamic changes that correspond to drug response or resistance, as has been observed for instance with RAS mutations in patients with colorectal cancer treated with epidermal growth factor receptor inhibitors [72].

4. How to Overcome Limitations: Functional Precision Medicine, Liquid Biopsy, and Molecular Tumor Board

4.1. Functional Precision Medicine: The Role of Patient-Derived Organoids (PDOs) and Patient-Derived Xenografts in a Personalized Approach

Functional precision cancer medicine has the potential to complement current genomic approaches. The minor role of molecular profiling in predicting response to targeted therapies and limitations of preclinical models currently used for drug selection have hindered correct validation of precision medicine [82]. The principal objective of functional models is the possibility to dynamically evaluate cancer evolution and clonal selection due to exposure to an anticancer drug. These models help us to grasp possible causes of treatment failure. During the last few years, 3D culture (the so-called organoids) and patient-derived xenograft models have both facilitated functional analysis for drug efficacy in solid tumors. Patient-derived xenograft (PDX) models have contributed in implementing translational medicine. These models are derived from the transplantation of patient tumor cells into immunodeficient mice. These models were found to reproduce in a more similar way the original tumor characteristics versus previous models in vivo. PDXs conserve the original tumor characteristics preserving the heterogeneity. PDXs could help in predicting drug sensitivity and resistance in several tumor types, being a good tool to improve personalized approach [83,84].
Derived from patient tumor cells, the organoids preserve tissue biological characteristics better than the more commonly used monolayer cell cultures. The possibility to test directly on the tumor of a specific patient represents a great step towards a personalized approach. In an intriguing recently published experiment, the PDOs analyzed showed 100% sensitivity, 93% specificity, 88% positive predictive value, and 100% negative predictive value in predicting patients’ response to treatment (Fisher’s exact test p < 0.0001). This marked a major breakthrough in precision medicine, suggesting that PDOs can contribute to treatment choice [85].

4.2. Dynamic Evaluation of Tumors: The Role of Liquid Biopsy

Another promising tool that has greatly facilitated dynamic evaluation of solid tumors is plasma cell-free DNA (cfDNA) analysis by liquid biopsy. Although a specific targetable biomarker can be identified, resistance always appears. For this reason, evaluation of tumor heterogeneity and clonal selection due to treatment pressure need to be deeply characterized. Several series have suggested that cfDNA could evaluate tumor heterogeneity by detecting the molecular mechanisms of acquired resistance [71,86]. This analysis allows genomics and other molecular alterations to be assessed at a specific moment, leading to the accurate evaluation of tumor development. In acquired resistance, the evolutionary pressure of therapy can drive outgrowth of distinct tumor sub-clones harboring independent resistance mechanisms within an individual patient, within different metastatic lesions, or even within the same lesion [87]. Moreover, the genomic analysis of a single biopsy upon disease progression may play a primary role in identifying mechanisms of acquired resistance. Nevertheless, this single-biopsy approach might not capture the massive heterogeneity of cancer cell populations shaped by the selective pressures of target therapy. cfDNA may offer advantages for assessing tumor heterogeneity. cfDNA analysis can potentially identify multiple concurrent heterogeneous resistance mechanisms that a single-biopsy approach could miss. In a recently published work, direct comparison of cfDNA versus tumor biopsy in the setting of acquired resistance illustrates how single-lesion tumor biopsies frequently fail to identify the presence of multiple clinically relevant resistance mechanisms, with cfDNA identifying additional concurrent resistance mechanisms in 78% of cases [88].

4.3. Molecular Tumor Board: Why Do We Need It?

The past few decades have seen a critical improvement in cancer treatment outcomes through a combination of molecular tumor testing and genetically matched targeted therapies. The history of precision medicine can be dated back to the first use of antiestrogen in breast cancer. Since this breakthrough, several other predictive biomarkers have been identified. Currently, the number of somatic tumor mutations used for standard treatment decisions is still quite limited and can easily be assessed via platforms capturing only these mutations, yet the demand for genetic testing evaluating molecular alterations will expand proportionally to rapidly expanding genetic knowledge and related drug development. Clinicians will be confronted with both increasingly complex genetic information and a wide array of platform choices; although large-scale sequencing is most informative in most cases, targeted in-depth sequencing may be preferred in others. The abundance of genetic tests and information provides a serious challenge. Furthermore, most pathology and sequencing reports only document well-known aberrations that could be therapeutically targeted, meaning that other genetic aberrations for which experimental agents and/or drug access programs are available may be missed [89]. Therefore, as novel molecular and genomic treatment indications are explored, it is becoming increasingly vital to generate and correctly interpret molecular tumor profiles to offer optimal cancer treatment. Nevertheless, the remarkable evolution of molecular techniques and the precision achieved by these tools need skilled assessment. This requires discussing the clinical cases and results obtained by molecular analyses to translate molecular profiles into clinical benefit for our patients. Developing molecular tumor boards made up of experts from a variety of disciplines will help optimize patient selection [90].

5. Conclusions

Personalized treatment for patients diagnosed with solid tumors has resulted in several advances in recent years. The possibility to offer a molecular-based personalized approach for cancer patients represents an attractive possibility in oncology. To obtain a relevant and real change which could improve all clinical outcome, a better understanding of molecular biology is needed. To improve, a multi-omic approach able to integrate DNA and RNA alterations, proteomics, and metabolomics will be necessary. The implementation of translational studies based on liquid biopsy and organoids or xenografts to evaluate molecular changes due to clonal pressure generated due to the use of target agents or tumor heterogeneity would help in the detection of mechanisms of resistance, suggesting the possibility for novel combinations. Further research is urgently needed to improve precision.

Funding

This research received no external funding.

Conflicts of Interest

A.C. declares institutional research funding from Genentech, Merck Serono, BMS, MSD, Roche, Beigene, Bayer, Servier, Lilly, Novartis, Takeda, Astellas and Fibrogen and advisory board or speaker fees from Merck Serono, Roche, Servier, Takeda and Astellas in the last 5 years. All remaining authors have declared no conflicts of interest.

References

  1. Berger, M.F.; Mardis, E.R. The emerging clinical relevance of genomics in cancer medicine. Nat. Rev. Clin. Oncol. 2018, 15, 353–365. [Google Scholar] [CrossRef]
  2. Lambert, A.W.; Pattabiraman, D.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef] [Green Version]
  3. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast cancer. Nat. Rev. Dis. Primers 2019, 5, 66. [Google Scholar] [CrossRef]
  4. Soverini, S.; Mancini, M.; Bavaro, L.; Cavo, M.; Martinelli, G. Chronic myeloid leukemia: The paradigm of targeting oncogenic tyrosine kinase signaling and counteracting resistance for successful cancer therapy. Mol. Cancer 2018, 17, 49. [Google Scholar] [CrossRef] [PubMed]
  5. Hyman, D.M.; Taylor, B.S.; Baselga, J. Implementing Genome-Driven Oncology. Cell 2017, 168, 584–599. [Google Scholar] [CrossRef] [PubMed]
  6. Ignatiadis, M.; Sotiriou, C. Luminal breast cancer: From biology to treatment. Nat. Rev. Clin. Oncol. 2013, 10, 494–506. [Google Scholar] [CrossRef] [PubMed]
  7. André, F.; Ciruelos, E.; Rubovszky, G.; Campone, M.; Loibl, S.; Rugo, H.S.; Iwata, H.; Conte, P.; Mayer, I.A.; Kaufman, B.; et al. Alpelisib for PIK3CA-Mutated, Hormone Receptor-Positive Advanced Breast Cancer. N. Engl. J. Med. 2019, 380, 1929–1940. [Google Scholar] [CrossRef] [PubMed]
  8. Swain, S.; Baselga, J.; Kim, S.-B.; Ro, J.; Semiglazov, V.; Campone, M.; Ciruelos, E.; Ferrero, J.-M.; Schneeweiss, A.; Heeson, S.; et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N. Engl. J. Med. 2015, 372, 724–734. [Google Scholar] [CrossRef] [Green Version]
  9. Finn, R.S.; Crown, J.P.; Lang, I.; Boer, K.; Bondarenko, I.M.; Kulyk, S.O.; Ettl, J.; Patel, R.; Pinter, T.; Schmidt, M.; et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): A randomised phase 2 study. Lancet Oncol. 2015, 16, 25–35. [Google Scholar] [CrossRef]
  10. Mok, T.S.; Kim, S.W.; Wu, Y.L.; Nakagawa, K.; Yang, J.J.; Ahn, M.J.; Wang, J.; Yang, J.C.H.; Lu, Y.; Atagi, S.; et al. Gefitinib Plus Chemotherapy Versus Chemotherapy in Epidermal Growth Factor Receptor Mutation-Positive Non-Small-Cell Lung Cancer Resistant to First-Line Gefitinib (IMPRESS): Overall Survival and Biomarker Analyses. J. Clin. Oncol. 2017, 35, 4027–4034. [Google Scholar] [CrossRef]
  11. Solomon, B.; Kim, N.-W.; Mekhail, T.; Paolini, J.; Usari, T.; Reisman, A.; Wilner, K.D.; Tursi, J.; Mok, T.S.; Wu, Y.-L.; et al. First-Line Crizotinib versus Chemotherapy in ALK-Positive Lung Cancer. N. Engl. J. Med. 2014, 371, 2167–2177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Rosell, R.; Carcereny, E.; Gervais, R.; Vergnenegre, A.; Massuti, B.; Felip, E.; Palmero, R.; Garcia-Gomez, R.; Pallares, C.; Sanchez, J.M.; et al. Erlotinib versus standard chemotherapy as fi rst-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): A multicentre, open-label, randomised phase 3 trial. Artic. Lancet Oncol. 2012, 13, 239–285. [Google Scholar] [CrossRef]
  13. Robert, C.; Grob, J.J.; Stroyakovskiy, D.; Karaszewska, B.; Hauschild, A.; Levchenko, E.; Sileni, V.C.; Schachter, J.; Garbe, C.; Bondarenko, I.; et al. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. N. Engl. J. Med. 2019, 381, 626–636. [Google Scholar] [CrossRef] [PubMed]
  14. Flaherty, K.T.; Puzanov, I.; Kim, K.B.; Ribas, A.; McArthur, G.A.; Sosman, J.A.; O’Dwyer, P.J.; Lee, R.J.; Grippo, J.F.; Nolop, K.; et al. Inhibition of Mutated, Activated BRAF in Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 809–819. [Google Scholar] [CrossRef] [Green Version]
  15. Hyman, D.M.; Piha-Paul, S.; Won, H.; Rodon, J.; Saura, C.; Shapiro, G.I.; Juric, D.; Quinn, D.I.; Moreno, V.; Doger, B.; et al. HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature 2018, 554, 189–194. [Google Scholar] [CrossRef]
  16. Smyth, L.M.; Piha-Paul, S.A.; Won, H.H.; Schram, A.M.; Saura, C.; Loi, S.; Lu, J.; Shapiro, G.I.; Juric, D.; Mayer, I.A.; et al. Efficacy and Determinants of Response to HER Kinase Inhibition in HER2-Mutant Metastatic Breast Cancer. Cancer Discov. 2019, 10, 198–213. [Google Scholar] [CrossRef] [Green Version]
  17. Krop, I.E.; Kim, S.-B.; González-Martín, A.; Lorusso, P.M.; Ferrero, J.-M.; Smitt, M.; Yu, R.; Leung, A.C.F.; Wildiers, H. Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): A randomised, open-label, phase 3 trial. Lancet Oncol. 2014, 15, 689–699. [Google Scholar] [CrossRef]
  18. Doi, T.; Shitara, K.; Naito, Y.; Shimomura, A.; Fujiwara, Y.; Yonemori, K.; Shimizu, C.; Shimoi, T.; Kuboki, Y.; Matsubara, N.; et al. Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody–drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: A phase 1 dose-escalation study. Lancet Oncol. 2017, 18, 1512–1522. [Google Scholar] [CrossRef]
  19. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef] [Green Version]
  20. Paez, J.G. EGFR Mutations in Lung Cancer: Correlation with Clinical Response to Gefitinib Therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef] [Green Version]
  21. Soda, M.; Choi, Y.L.; Enomoto, M.; Takada, S.; Yamashita, Y.; Ishikawa, S.; Fujiwara, S.-I.; Watanabe, H.; Kurashina, K.; Hatanaka, H.; et al. Identification of the transforming EML4–ALK fusion gene in non-small-cell lung cancer. Nature 2007, 448, 561–566. [Google Scholar] [CrossRef] [PubMed]
  22. Borger, D.R.; Tanabe, K.K.; Fan, K.C.; Lopez, H.U.; Fantin, V.R.; Straley, K.S.; Schenkein, D.P.; Hezel, A.F.; Ancukiewicz, M.; Liebman, H.M.; et al. Frequent Mutation of Isocitrate Dehydrogenase (IDH)1 and IDH2 in Cholangiocarcinoma Identified Through Broad-Based Tumor Genotyping. Oncology 2011, 17, 72–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [PubMed]
  24. Adashek, J.J.; Lorusso, P.M.; Hong, D.S.; Kurzrock, R. Phase I trials as valid therapeutic options for patients with cancer. Nat. Rev. Clin. Oncol. 2019, 16, 773–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Gonzalez-Angulo, A.M.; Timms, K.M.; Liu, S.; Chen, H.; Litton, J.K.; Potter, J.; Lanchbury, J.S.; Stemke-Hale, K.; Hennessy, B.T.; Arun, B.K.; et al. Incidence and outcome of BRCA mutations in unselected patients with triple receptor-negative breast cancer. Clin. Cancer Res. 2011, 17, 1082–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Alsop, K.; Fereday, S.; Meldrum, C.; DeFazio, A.; Emmanuel, C.; George, J.; Dobrovic, A.; Birrer, M.J.; Webb, P.; Stewart, C.; et al. BRCA Mutation Frequency and Patterns of Treatment Response in BRCA Mutation–Positive Women with Ovarian Cancer: A Report from the Australian Ovarian Cancer Study Group. J. Clin. Oncol. 2012, 30, 2654–2663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Dienstmann, R.; Rodon, J.; Prat, A.; Perez-Garcia, J.; Adamo, B.; Felip, E.; Cortés, J.; Iafrate, A.J.; Nuciforo, P.; Tabernero, J. Genomic aberrations in the FGFR pathway: Opportunities for targeted therapies in solid tumors. Ann. Oncol. 2014, 25, 552–563. [Google Scholar] [CrossRef]
  28. Drilon, A.; Laetsch, T.W.; Kummar, S.; Dubois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.; et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef]
  29. Kimmelman, J.; Tannock, I. The paradox of precision medicine. Nat. Rev. Clin. Oncol. 2018, 15, 341–342. [Google Scholar] [CrossRef]
  30. Rodriguez, H.; Pennington, S.R. Revolutionizing Precision Oncology through Collaborative Proteogenomics and Data Sharing. Cell 2018, 173, 535–539. [Google Scholar] [CrossRef] [Green Version]
  31. Mertins, P.; Mani, D.R.; Ruggles, K.V.; Gillette, M.A.; Clauser, K.R.; Wang, P.; Wang, X.; Qiao, J.W.; Cao, S.; Petralia, F.; et al. Proteogenomics connects somatic mutations to signaling in breast cancer. Nature 2016, 534, 55–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Puchades-Carrasco, L.; Lucena, A.P. Metabolomics Applications in Precision Medicine: An Oncological Perspective. Curr. Top. Med. Chem. 2017, 17, 2740–2751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Clayton, T.A.; Lindon, J.C.; Cloarec, O.; Antti, H.; Charuel, C.; Hanton, G.; Provost, J.P.; Le Net, J.L.; Baker, D.; Walley, R.J.; et al. Pharmaco-metabonomic phenotyping and personalized drug treatment. Nature 2006, 440, 1073–1077. [Google Scholar] [CrossRef] [PubMed]
  34. Gambardella, V.; Gimeno-Valiente, F.; Tarazona, N.; Martínez-Ciarpaglini, C.; Roda, D.; Fleitas, T.; Tolosa, P.; Cejalvo, J.M.; Huerta, M.; Roselló, S.; et al. NRF2 through RPS6 Activation Is Related to Anti-HER2 Drug Resistance in HER2-Amplified Gastric Cancer. Clin. Cancer Res. 2018, 25, 1639–1649. [Google Scholar] [CrossRef]
  35. Rojo de Vega, M.; Chapman, E.; Zhang, D.D. NRF2 and the hallmarks of cáncer. Cancer Cell 2019, 34, 21–43. [Google Scholar] [CrossRef]
  36. Garralda, E.; Dienstmann, R.; Piris-Giménez, A.; Braña, I.; Rodon, J.; Tabernero, J. New clinical trial designs in the era of precision medicine. Mol. Oncol. 2019, 13, 549–557. [Google Scholar] [CrossRef]
  37. Schwaederle, M.; Zhao, M.; Lee, J.J.; Eggermont, A.M.; Schilsky, R.L.; Mendelsohn, J.; Lazar, V.; Kurzrock, R. Impact of Precision Medicine in Diverse Cancers: A Meta-Analysis of Phase II Clinical Trials. J. Clin. Oncol. 2015, 33, 3817–3825. [Google Scholar] [CrossRef]
  38. Jardim, D.L.; Schwaederle, M.; Wei, C.; Lee, J.J.; Hong, D.S.; Eggermont, A.M.; Schilsky, R.L.; Mendelsohn, J.; Lazar, V.; Kurzrock, R. Impact of a Biomarker-Based Strategy on Oncology Drug Development: A Meta-Analysis of Clinical Trials Leading to FDA Approval. J. Natl. Cancer Inst. 2015, 107. [Google Scholar] [CrossRef] [Green Version]
  39. Massard, C.; Michiels, S.; Ferté, C.; Le Deley, M.-C.; Lacroix, L.; Hollebecque, A.; Verlingue, L.; Ileana, E.; Rosellini, S.; Ammari, S.; et al. High-Throughput Genomics and Clinical Outcome in Hard-to-Treat Advanced Cancers: Results of the MOSCATO 01 Trial. Cancer Discov. 2017, 7, 586–595. [Google Scholar] [CrossRef] [Green Version]
  40. Goncalves, M.D.; Hopkins, B.D.; Cantley, L.C. Phosphatidylinositol 3-Kinase, Growth Disorders, and Cancer. N. Engl. J. Med. 2018, 379, 2052–2062. [Google Scholar] [CrossRef]
  41. Hong, R.; Edgar, K.; Song, K.; Steven, S.; Young, A.; Hamilton, P.; Arrazate, A.; De La Cruz, C.; Chan, C.; Pang, J.; et al. Abstract PD4-14: GDC-0077 is a selective PI3Kalpha inhibitor that demonstrates robust efficacy in PIK3CA mutant breast cancer models as a single agent and in combination with standard of care therapies. Cancer Res. 2018, 78, PD4-14. [Google Scholar]
  42. Merlino, G.; Fiascarelli, A.; Bigioni, M.; Bressan, A.; Irrissuto, C.; Pellacani, A.; Scaltriti, M.; Binaschi, M. Abstract 2160: MEN1611, a novel α-selective PI3K inhibitor in solid tumors. Tumor Biol. 2018, 78, 2160. [Google Scholar] [CrossRef]
  43. Ottensmeier, C.H.; Jones, T.; Sacco, J.J.; McCaul, J.; Brennan, P.; Paterson, C.; Schache, A.; Shaw, R.J.; Singh, R.P.; Davies, J.H.; et al. A randomised, double-blind, placebo-controlled phase IIa trial of AMG319 given orally as neoadjuvant therapy in patients with human papillomavirus (HPV) positive and negative head and neck squamous cell carcinoma (HNSCC). J. Clin. Oncol. 2018, 36, 6068. [Google Scholar] [CrossRef]
  44. Blagden, S.; Olmin, A.; Josephs, D.; Stavraka, C.; Zivi, A.; Pinato, D.J.; Anthoney, A.; Decordova, S.; Swales, K.; Riisnaes, R.; et al. Correction: First-in-human study of CH5132799, an oral class I PI3K inhibitor, studying toxicity, pharmacokinetics, and pharmacodynamics, in patients with metastatic cancer. Clin. Cancer Res. 2015, 21, 660. [Google Scholar]
  45. Paik, P.K.; Shen, R.; Berger, M.F.; Ferry, D.; Soria, J.-C.; Mathewson, A.; Rooney, C.; Smith, N.R.; Cullberg, M.; Kilgour, E.; et al. A Phase Ib Open-Label Multicenter Study of AZD4547 in Patients with Advanced Squamous Cell Lung Cancers. Clin. Cancer Res. 2017, 23, 5366–5373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Pal, S.K.; Rosenberg, J.E.; Hoffman-Censits, J.H.; Berger, R.; Quinn, D.I.; Galsky, M.D.; Wolf, J.; Dittrich, C.; Keam, B.; Delord, J.-P.; et al. Efficacy of BGJ398, a Fibroblast Growth Factor Receptor 1-3 Inhibitor, in Patients with Previously Treated Advanced Urothelial Carcinoma with FGFR3 Alterations. Cancer Discov. 2018, 8, 812–821. [Google Scholar] [CrossRef] [Green Version]
  47. Koyama, T.; Shimizu, T.; Iwasa, S.; Fujiwara, Y.; Kondo, S.; Kitano, S.; Yonemori, K.; Shimomura, A.; Iizumi, S.; Sasaki, T.; et al. Abstract B160: First-in-human phase 1 study of E7090, a novel selective inhibitor of FGFRs, in patients with advanced solid tumors. Mol. Cancer Ther. 2018, 17, B160. [Google Scholar] [CrossRef]
  48. Michael, M.; Bang, Y.-J.; Park, Y.S.; Kang, Y.-K.; Kim, T.M.; Hamid, O.; Thornton, N.; Tate, S.C.; Raddad, E.; Tie, J. A Phase 1 Study of LY2874455, an Oral Selective pan-FGFR Inhibitor, in Patients with Advanced Cancer. Target. Oncol. 2017, 12, 463–474. [Google Scholar] [CrossRef]
  49. Goyal, L.; Shi, L.; Liu, L.Y.; De La Cruz, F.F.; Lennerz, J.K.; Raghavan, S.; Leschiner, I.; Elagina, L.; Siravegna, G.; Ng, R.; et al. TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion-Positive Intrahepatic Cholangiocarcinoma. Cancer Discov. 2019, 9, 1064–1079. [Google Scholar] [CrossRef] [Green Version]
  50. Kim, R.D.; Sarker, D.; Meyer, T.; Yau, T.; Macarulla, T.; Park, J.-W.; Choo, S.P.; Hollebecque, A.; Sung, M.W.; Lim, H.-Y.; et al. First-in-Human Phase I Study of Fisogatinib (BLU-554) Validates Aberrant FGF19 Signaling as a Driver Event in Hepatocellular Carcinoma. Cancer Discov. 2019, 9, 1696–1707. [Google Scholar] [CrossRef] [Green Version]
  51. Mercade, T.M.; Moreno, V.; John, B.; Morris, J.C.; Sawyer, M.B.; Yong, W.P.; Gutierrez, M.; Karasic, T.B.; Sangro, B.; Sheng-Shun, Y.; et al. A phase I study of H3B-6527 in hepatocellular carcinoma (HCC) or intrahepatic cholangiocarcinoma (ICC) patients (pts). J. Clin. Oncol. 2019, 37, 4095. [Google Scholar] [CrossRef]
  52. Weiss, A.; Adler, F.; Buhles, A.; Stamm, C.; Fairhurst, R.A.; Kiffe, M.; Sterker, D.; Centeleghe, M.; Wartmann, M.; Kinyamu-Akunda, J.; et al. FGF401, A First-In-Class Highly Selective and Potent FGFR4 Inhibitor for the Treatment of FGF19-Driven Hepatocellular Cancer. Mol. Cancer Ther. 2019, 18, 2194–2206. [Google Scholar] [CrossRef] [PubMed]
  53. Hyman, D.; Kummar, S.; Farago, A.; Geoerger, B.; Mau-Sorensen, M.; Taylor, M.; Garralda, E.; Nagasubramanian, R.; Natheson, M. Abstract CT127: Phase I and expanded access experience of LOXO-195 (BAY 2731954), a selective next-generation TRK inhibitor (TRKi). Cancer Res. 2019, 79, CT127. [Google Scholar]
  54. Lin, C.-C.; Arkenau, H.-T.; Lu, S.; Sachdev, J.; Carpeño, J.D.C.; Mita, M.; Dziadziuszko, R.; Su, W.-C.; Bobilev, D.; Hughes, L.; et al. A phase 1, open-label, dose-escalation trial of oral TSR-011 in patients with advanced solid tumours and lymphomas. Br. J. Cancer 2019, 121, 131–138. [Google Scholar] [CrossRef] [PubMed]
  55. Papadopoulos, K.P.; Gandhi, L.; Jänne, P.A.; Ou, S.-H.I.; Shaw, A.; Goldberg, T.R.; Greenberg, J.; Gu, X.; Tachibana, M.; Senaldi, G.; et al. First-in-human study of DS-6051b in patients (pts) with advanced solid tumors (AST) conducted in the US. J. Clin. Oncol. 2018, 36, 2514. [Google Scholar] [CrossRef]
  56. Dieci, M.; Arnedos, M.; André, F.; Soria, J.-C. Fibroblast Growth Factor Receptor Inhibitors as a Cancer Treatment: From a Biologic Rationale to Medical Perspectives. Cancer Discov. 2013, 3, 264–279. [Google Scholar] [CrossRef] [Green Version]
  57. Babina, I.; Turner, N.C. Advances and challenges in targeting FGFR signalling in cancer. Nat. Rev. Cancer 2017, 17, 318–332. [Google Scholar] [CrossRef]
  58. Siefker-Radtke, A.; Curti, B. Immunotherapy in metastatic urothelial carcinoma: Focus on immune checkpoint inhibition. Nat. Rev. Urol. 2017, 15, 112–124. [Google Scholar] [CrossRef]
  59. Loriot, Y.; Necchi, A.; Park, S.H.; Garcia-Donas, J.; Huddart, R.; Burgess, E.F.; Fleming, M.; Rezazadeh, A.; Mellado, B.; Varlamov, S.; et al. Erdafitinib in Locally Advanced or Metastatic Urothelial Carcinoma. N. Engl. J. Med. 2019, 381, 338–348. [Google Scholar] [CrossRef]
  60. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
  61. Yaeger, R.; Chatila, W.K.; Lipsyc, M.D.; Hechtman, J.F.; Cercek, A.; Sanchez-Vega, F.; Jayakumaran, G.; Middha, S.; Zehir, A.; Donoghue, M.T.A.; et al. Clinicla sequencing defines the genomic landscape of metastatic colorectal cancer. Cancer Cell 2019, 33, 125–136. [Google Scholar] [CrossRef] [PubMed]
  62. Kopetz, S.; Grothey, A.; Yaeger, R.; Van Cutsem, E.; Desai, J.; Yoshino, T.; Wasan, H.; Ciardiello, F.; Loupakis, F.; Hong, Y.S.; et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E-Mutated Colorectal Cancer. N. Engl. J. Med. 2019, 381, 1632–1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Märkl, B.; Hirschbühl, K.; Dhillon, C. NTRK-Fusions—A new kid on the block. Pathol. Res. Pract. 2019, 215, 152572. [Google Scholar] [CrossRef] [PubMed]
  64. Le Tourneau, C.; Delord, J.-P.; Gonçalves, A.; Gavoille, C.; Dubot, C.; Isambert, N.; Campone, M.; Trédan, O.; Massiani, M.-A.; Mauborgne, C.; et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): A multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015, 16, 1324–1334. [Google Scholar] [CrossRef]
  65. Barroilhet, L.; Matulonis, U. The NCI-MATCH trial and precision medicine in gynecologic cancers. Gynecol. Oncol. 2018, 148, 585–590. [Google Scholar] [CrossRef] [PubMed]
  66. Dagogo-Jack, I.; Shaw, A.T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 2017, 15, 81–94. [Google Scholar] [CrossRef]
  67. Eckhardt, S.G.; Lieu, C. Is Precision Medicine an Oxymoron? JAMA Oncol. 2019, 5, 142. [Google Scholar] [CrossRef]
  68. Gambardella, V.; Fleitas, T.; Tarazona, N.; Cejalvo, J.; Gimeno-Valiente, F.; Martínez-Ciarpaglini, C.; Huerta, M.; Roselló, S.; Castillo, J.; Roda, D.; et al. Towards precision oncology for HER2 blockade in gastroesophageal adenocarcinoma. Ann. Oncol. 2019, 30, 1254–1264. [Google Scholar] [CrossRef] [Green Version]
  69. Polasek, T.M.; Shakib, S.; Rostami-hodjegan, A. Expert Review of Clinical Pharmacology Precision dosing in clinical medicine: Present and future. Expert Rev. Clin. Pharmacol. 2018, 11, 743–746. [Google Scholar] [CrossRef] [Green Version]
  70. Darwich, A.S.; Ogungbenro, K.; Vinks, A.A.; Powell, J.R.; Reny, J.L.; Marsousi, N.; Daali, Y.; Fairman, D.; Cook, J.; Lesko, L.J.; et al. Why has model-informed precision dosing not yet become common clinical reality? Lessons from the past and a roadmap for the future. Clin. Pharmacol. Ther. 2017, 101, 646–656. [Google Scholar] [CrossRef]
  71. McGranahan, N.; Swanton, C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 2015, 27, 15–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Diaz, L.A.; Bardelli, A. Liquid Biopsies: Genotyping Circulating Tumor DNA. J. Clin. Oncol. 2014, 32, 579–586. [Google Scholar] [CrossRef] [PubMed]
  73. Birkbak, N.J.; Mcgranahan, N. Review Cancer Genome Evolutionary Trajectories in Metastasis. Cancer Cell 2019, 37, 8–19. [Google Scholar] [CrossRef] [PubMed]
  74. El-Deiry, W.S.; Goldberg, R.M.; Lenz, H.; Shields, A.F.; Gibney, G.T.; Tan, A.R.; Brown, J.; Eisenberg, B.; Heath, E.I.; Phuphanich, S.; et al. The current state of molecular testing in the treatment of patients with solid tumors, 2019. CA A Cancer J. Clin. 2019, 69, 305–343. [Google Scholar] [CrossRef] [PubMed]
  75. Von Hoff, D.D.; Stephenson, J.J.; Rosen, P.; Loesch, D.M.; Borad, M.J.; Anthony, S.; Jameson, G.S.; Brown, S.; Cantafio, N.; Richards, D.; et al. Pilot Study Using Molecular Profiling of Patients’ Tumors to Find Potential Targets and Select Treatments for Their Refractory Cancers. J. Clin. Oncol. 2010, 28, 4877–4883. [Google Scholar] [CrossRef] [PubMed]
  76. Tsimberidou, A.M.; Iskander, N.G.; Hong, D.S.; Wheler, J.J.; Falchook, G.S.; Fu, S.; Piha-Paul, S.; Naing, A.; Janku, F.; Luthra, R.; et al. Personalized medicine in a phase I clinical trials program: The MD Anderson Cancer Center initiative. Clin. Cancer Res. 2012, 18, 6373–6383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Hainsworth, J.D.; Meric-Bernstam, F.; Swanton, C.; Hurwitz, H.; Spigel, D.R.; Sweeney, C.; Burris, H.A.; Bose, R.; Yoo, B.; Stein, A.; et al. Targeted Therapy for Advanced Solid Tumors on the Basis of Molecular Profiles: Results from MyPathway, an Open-Label, Phase IIa Multiple Basket Study. J. Clin. Oncol. 2018, 36, 536–542. [Google Scholar] [CrossRef]
  78. Trédan, O.; Wang, Q.; Pissaloux, D.; Cassier, P.; De La Fouchardière, A.; Fayette, J.; Desseigne, F.; Ray-Coquard, I.; Frappaz, D.; Heudel, P.-E.; et al. Molecular screening program to select molecular-based recommended therapies for metastatic cancer patients: Analysis from the ProfiLER trial. Ann. Oncol. 2019, 30, 757–765. [Google Scholar] [CrossRef]
  79. Sicklick, J.K.; Kato, S.; Okamura, R.; Schwaederle, M.; Hahn, M.E.; Williams, C.B.; De, P.; Krie, A.; Piccioni, D.E.; Miller, V.A.; et al. Molecular profiling of cancer patients enables personalized combination therapy: The I-PREDICT study. Nat. Med. 2019, 25, 744–750. [Google Scholar] [CrossRef]
  80. Rodon, J.; Soria, J.-C.; Berger, R.; Miller, W.H.; Rubin, E.; Kugel, A.; Tsimberidou, A.; Saintigny, P.; Ackerstein, A.; Braña, I.; et al. Genomic and transcriptomic profiling expands precision cancer medicine: The WINTHER trial. Nat. Med. 2019, 25, 751–758. [Google Scholar] [CrossRef]
  81. Clevers, H. Modeling Development and Disease with Organoids. Cell 2016, 165, 1586–1597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Letai, A. Functional precision cancer medicine—Moving beyond pure genomics. Nat. Med. 2017, 23, 1028–1035. [Google Scholar] [CrossRef] [PubMed]
  83. Okada, S.; Vaeteewoottacharn, K.; Kariya, R. Establishment of a Patient-Derived Tumor Xenograft Model and Application for Precision Cancer Medicine. Chem. Pharm. Bull. 2018, 66, 225–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Bhimani, J.; Ball, K.; Stebbing, J. Patient-derived xenograft models—The future of personalised cancer treatment. Br. J. Cancer 2020, 122, 601–602. [Google Scholar] [CrossRef]
  85. Vlachogiannis, G.; Hedayat, S.; Vatsiou, A.; Jamin, Y.; Fernández-Mateos, J.; Khan, K.; Lampis, A.; Eason, K.; Huntingford, I.; Burke, R.; et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018, 359, 920–926. [Google Scholar] [CrossRef] [Green Version]
  86. Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of Circulating Tumor DNA in Early- and Late-Stage Human Malignancies. Sci. Transl. Med. 2014, 6, 224ra24. [Google Scholar] [CrossRef] [Green Version]
  87. Gerlinger, M.; Rowan, A.J.; Horswell, S.; Math, M.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; et al. Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing. N. Engl. J. 2012, 366, 883–892. [Google Scholar] [CrossRef] [Green Version]
  88. Parikh, A.R.; Leshchiner, I.; Elagina, L.; Goyal, L.; Levovitz, C.; Siravegna, G.; Livitz, D.; Rhrissorrakrai, K.; Martin, E.E.; Van Seventer, E.E.; et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat. Med. 2019, 25, 1415–1421. [Google Scholar] [CrossRef]
  89. Van Der Velden, D.; Van Herpen, C.; Van Laarhoven, H.; Smit, E.; Groen, H.; Willems, S.; Nederlof, P.; Langenberg, M.; Cuppen, E.; Sleijfer, S.; et al. Molecular Tumor Boards: Current practice and future needs. Ann. Oncol. 2017, 28, 3070–3075. [Google Scholar] [CrossRef]
  90. Van De Haar, J.; Hoes, L.; Voest, E. Advancing molecular tumour boards: Highly needed to maximise the impact of precision medicine. ESMO Open 2019, 4, e000516. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Personalized treatment: an integrated precision approach. How to personalize cancer treatment from the molecular evaluation of primary tumor or metastases evaluating liquid biopsy, tumor-derived organoids, and tumor-derived xenografts.
Figure 1. Personalized treatment: an integrated precision approach. How to personalize cancer treatment from the molecular evaluation of primary tumor or metastases evaluating liquid biopsy, tumor-derived organoids, and tumor-derived xenografts.
Cancers 12 01009 g001
Figure 2. Molecular alterations driving precision oncology.
Figure 2. Molecular alterations driving precision oncology.
Cancers 12 01009 g002
Table 1. Novel targeted agents against PIK3CA1, FGFR2, and NTRK3 molecular alterations under development for solid tumors in phase I trials.
Table 1. Novel targeted agents against PIK3CA1, FGFR2, and NTRK3 molecular alterations under development for solid tumors in phase I trials.
Class of InhibitorsNovel Targeted Agents under Development for Solid Tumors in Phase I Trials
Compound NameMechanism of ActionPhase
PI3K inhibitorsGDC0077 [41]Potent PI3K alpha inhibitorIb
MEN1611 (PA799) [42]PI3K alpha inhibitorIb
AMG319 [43]AMG319 is a PI3Kδ inhibitor. Preclinically, target inhibition abrogates Treg-mediated immunosuppression, augmenting CD8+ T-cell antitumor activityIIa
CH5132799 [44]Oral pan-PI3 kinase inhibitorIa/b
FGFR inhibitorsAZD4547 [45]Potent and selective inhibitor of FGFR 1, 2, and 3I
NVP-BGJ398 [46]Oral, selective, ATP-competitive inhibitor of FGFR1, 2, and 3I
E-7090 [47]Oral and selective inhibitor of FGFR1, 2, and 3I
LY2874455 [48]Inhibitor of FGFR 1, 2, 3, and 4I
TAS-120 [49]Potent and highly specific against wildtype FGFR1–4 as well as against some FGFR2 kinase domain mutations I
BLU-554 [50]Potent and selective inhibitor of FGFR4I
H3B-6527 [51]Selective and covalent inhibitor of FGFR4I
FGF-401 [52]Potent and selective, reversible-covalent small-molecule inhibitor of FGFR4I
NTRK inhibitorLOXO-195 [53]Selective inhibitor of TRKI
TSR-011 [54]Dual ALK4 and TRK inhibitorI
DS-6051b [55]Inhibitor with high affinity for ROS1 5 and TRKI
1 PIK3CA = Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha; FGFR 2 = Fibroblast Growth Factor Receptor; NTRK 3 = Neurotrophic tropomyosin receptor kinas; ALK 4 = Anaplastic lymphoma kinase; ROS1 5 = c-ros oncogene 1.
Table 2. Molecular tools for selecting patients in clinical trials.
Table 2. Molecular tools for selecting patients in clinical trials.
Molecular Tools for Selecting Patients in Precision Medicine-Based Basket Trials
Clinical TrialMolecular Tools
Bisgrove [75]Immunohistochemistry, Fluorescence in situ hybridization microarray
IMPACT [76]PCR-based genomics and NGS
SHIVA [64]Targeted NGS-based
MOSCATO [39]Targeted NGS-based, RNA Seq
MyPathway [77]Genomic testing
Profiler [78]Targeted NGS-based
I-PREDICT [79]Targeted NGS-based, ctDNA
WINTHER [80]Targeted NGS-based, Transcriptomic

Share and Cite

MDPI and ACS Style

Gambardella, V.; Tarazona, N.; Cejalvo, J.M.; Lombardi, P.; Huerta, M.; Roselló, S.; Fleitas, T.; Roda, D.; Cervantes, A. Personalized Medicine: Recent Progress in Cancer Therapy. Cancers 2020, 12, 1009. https://doi.org/10.3390/cancers12041009

AMA Style

Gambardella V, Tarazona N, Cejalvo JM, Lombardi P, Huerta M, Roselló S, Fleitas T, Roda D, Cervantes A. Personalized Medicine: Recent Progress in Cancer Therapy. Cancers. 2020; 12(4):1009. https://doi.org/10.3390/cancers12041009

Chicago/Turabian Style

Gambardella, Valentina, Noelia Tarazona, Juan Miguel Cejalvo, Pasquale Lombardi, Marisol Huerta, Susana Roselló, Tania Fleitas, Desamparados Roda, and Andres Cervantes. 2020. "Personalized Medicine: Recent Progress in Cancer Therapy" Cancers 12, no. 4: 1009. https://doi.org/10.3390/cancers12041009

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop