Skip to main content
  • Research highlight
  • Open access
  • Published:

Basket trial of TRK inhibitors demonstrates efficacy in TRK fusion-positive cancers

Abstract

Unlike many conventional cancers with preferential patterns of oncogenic genetic alterations, TRK fusions resulting from NTRK1/2/3 genetic alterations drive oncogenic transformations in more than 20 different malignancies over diverse tissue/cell lineages, in both children and adults. A recent “basket” study of larotrectinib, a TRK inhibitor, has demonstrated significant efficacy in TRK fusion-positive tumors of all types from infants to the elderly. Here, we discuss the larotrectinib study and perspectives and challenges in developing “tumor-agnostic” targeted therapies in rare tumors.

Background

Traditionally, cancers are classified and treated based on their pathologic classification and tissue of origin. Advances in sequencing technology and large-scale cancer genomics effort (e.g., the International Cancer Genome Consortium and The Cancer Genome Atlas (TCGA) have identified many targetable driver genetic alterations across different tumor types and have shifted the cancer sub-classification based on driver genetic alterations. For example, lung adenocarcinomas are further sub-classified by KRAS and EGFR mutations and ALK and ROS1 translocation. Most “driver” genetic alterations are preferentially found in certain cell/tissue lineages. For example, the BRAFV600 mutation occurs at high frequencies in specific tumor types, e.g., melanoma, thyroid cancer, hairy cell leukemia, Langerhans cell histiocytosis, and colorectal cancer, and at significantly lower frequencies in other tissue lineages. The responses to BRAFV600-targeted therapies are not uniform, with some cancer types (e.g., colorectal cancer) exhibit tissue lineage-specific primary resistance [1,2,3], underlining the importance of the tissue lineage-specific cellular context.

TRK fusions are rare but drive oncogenesis in diverse tissue lineages

The NTRK1, NTRK2, and NTRK3 genes, encoding the tropomyosin receptor kinases (TRK), TRKA, TRKB, and TRKC, respectively, are receptor tyrosine kinases that are normally expressed in the nervous system [4]. Physiologically, TRK receptor tyrosine kinases are activated by binding of mature neurotrophins, which mediate neuronal survival and synaptic plasticity in the central nervous system [4]. NTRK genetic alterations (e.g., translocations) resulting in TRK fusion proteins can lead to ligand-independent activation of TRK kinases and drive oncogenic transformation [5,6,7]. To date, TRK fusions are found in more than 20 different tumor types. With the exception of several rare tumor types (e.g., secretory breast carcinoma, mammary analog secretory carcinoma, congenital fibrosarcomas, and congenital mesoblastic nephroma), the majority of the TRK fusions occur in low frequencies in a variety of common cancers over a diverse tissue/cell lineages (e.g., lung adenocarcinoma, sarcoma, acute myeloid leukemia, colorectal cancer) [6, 7] (Table 1). The rarity of TRK fusions and the heterogeneity of tumor types present incredible challenges to clinically evaluate TRK inhibitors. Diagnostically, because of large introns, these fusions are difficult to detect using multiplex targeted exome capture panels (e.g., FoundationOne®, MSK-IMPACT™).

Table 1 Rare TRK fusions in diverse tumor types

TRK inhibitor larotrectinib demonstrates efficacy in a basket trial

Recently, Drilon and colleagues reported a phase I/II clinical trial to evaluate the safety and efficacy of larotrectinib, a highly selective small-molecule inhibitor of all three TRK proteins, using a novel “basket” trial design that enrolled patients based on NTRK genetic alterations regardless of age or tumor types [8]. A total of 55 patients (ages 4 months–76 years old) with 16 different tumor histologies were treated on three protocols, and the results were pooled. The investigators found that larotrectinib was generally well-tolerated with < 5% treatment-related grade 3 or 4 adverse events. The overall RECIST response rate was 75% (95% confidence interval, 61–85) by independent review and 80% (95% confidence interval, 67–90) by investigator assessment. At 1 year, 71% of patients are with ongoing responses and 55% of patients remain progression-free. The median duration of response and progression-free survival has not been reached after 8.3 and 9.9 months of median follow-up, respectively. Importantly, responses were observed in nearly all tumor types and age groups. Three of the six patients who did not response to larotrectinib (primary resistance) had undetectable TRK proteins by immunohistochemistry (IHC) despite molecularly identified TRK fusion at a screening in local laboratories. In the ten patients who progressed after an initial response for at least 6 months, nine had identifiable secondary resistant mutations in NTRK1 or NTRK3, including substitutions in the solvent front position (NTRK1 G595R or NTRK3 G623R), gatekeeper mutation (NRTK1 F589 L), and the xDFG position NTRK1 G667S or NTRK3 G696A). The acquired resistance mechanisms have been described for other oncogenic kinase-targeted therapies [9,10,11]. The next generation of TRK inhibitors is in development to overcome the acquired resistance in TRK [7, 12] (see Table 2).

Table 2 TRK inhibitors currently in clinical development

Future perspectives

The study by Drillon et al. [8] comes on the heels of several basket trials, including the AKT inhibitor AZD5363 in AKT1 E17K-mutant tumors [13], the PD1 inhibitor pembrolizumab in mismatch repair deficient tumors [14], and the pan-HER kinase inhibitor neratinib in HER2- and HER3-mutant tumors [15], with variable clinical success. This current study provides a compelling case for tumor-agnostic, molecular-driven “basket” approaches for clinical investigations of rare driver mutations across diverse tumor types. It paves a clinical pathway to effective therapeutics for patients with rare tumors and rare driver mutations. In addition to larotrectinib, there is a variety of TRK inhibitors currently in clinical development (Table 1), including next-generation TRK inhibitors that can overcome acquired resistance (e.g., LOXO-195, TPX-0005).

Despite the early clinical success with new generations of TRK inhibitors and novel trial design, the challenges remain for real-time identification of rare TRK fusions. What would be the ideal diagnostics methodology? DNA-based next-generation sequencing (NGS) assays have relatively high false-negative and false-positive rate and do not identify novel fusions. RNA-based NGS assays (e.g., Archer Dx) can detect novel fusions and has reasonable sensitivity. However, both DNA- and RNA-based NGS assays can be costly and effort intensive. Alternatively, IHC of TRK is a sensitive and efficient method for identification of TRK expression [16, 17]. Nevertheless, it would not readily discriminate TRK fusion arising through genetic alterations where TRK inhibitors can be highly effective from full-length TRK expression in tumors inherited through development where the functional significance of TRK expression and clinically impact is unknown. TRK IHC can also be associated with false positives in certain tissue and tumor types. Furthermore, unlike NGS-based assays, IHC cannot be easily multiplexed into a panel without added cost and effort. While TRK IHC can be easily justified for high-prevalence tumors (e.g., congenital fibrosarcoma or secretory breast carcinoma), its role in low-prevalence common tumors such as colorectal cancer becomes more debatable. Importantly, it is unclear which diagnostic modality, NGS of NTRK alterations or IHC of TRK expression, is more predictive of response to TRK inhibitors. In the NEJM by Drilon et al., three out of the six non-responders to larotrectinib did not have centrally confirmed TRK expression by pan-TRK IHC, despite the detection of NTRK rearrangement by NGS in the local laboratory [8]. This observation suggests that TRK expression by IHC may be necessary for response. Currently, Pan-TRK IHC and Illumina NGS (RNA and DNA assays in one design) are both being developed as companion diagnostics to larotrectinib and other TRK inhibitors.

With a shifting paradigm of identifying genetic alterations in a tumor-agnostic manner, the development of a single assay that can identify multiple types of actionable genetic alterations would be paramount. In the meantime, TRK IHC would be a reasonable initial diagnostics for rare tumors where TRK fusions are frequent, and possibly common tumors where driver mutations are absent.

References

  1. Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med. 2015;373:726–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Prahallad A, Sun C, Huang S, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100–3.

    Article  PubMed  CAS  Google Scholar 

  3. Corcoran RB, Ebi H, Turke AB, et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discovery. 2012;2:227–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Chao MV. Neurotrophins and their receptors: a convergence point for many signalling pathways. Nat Rev Neurosci. 2003;4:299–309.

    Article  PubMed  CAS  Google Scholar 

  5. Martin-Zanca D, Hughes SH, Barbacid M. A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature. 1986;319:743–8.

    Article  PubMed  CAS  Google Scholar 

  6. Vaishnavi A, Le AT, Doebele RC. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discovery. 2015;5:25–34.

    Article  PubMed  CAS  Google Scholar 

  7. Amatu A, Sartore-Bianchi A, Siena S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open. 2016;1:e000023.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med. 2018;378:731–9.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  9. Katayama R, Shaw AT, Khan TM, et al. Mechanisms of acquired crizotinib resistance in ALK-rearranged lung cancers. Sci Transl Med. 2012;4:120ra17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Kobayashi S, Boggon TJ, Dayaram T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med. 2005;352:786–92.

    Article  PubMed  CAS  Google Scholar 

  11. Core LJ, Martins AL, Danko CG, Waters CT, Siepel A, Lis JT. Analysis of nascent RNA identifies a unified architecture of initiation regions at mammalian promoters and enhancers. Nat Genet. 2014;46:1311–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Drilon A, Nagasubramanian R, Blake JF, et al. A next-generation TRK kinase inhibitor overcomes acquired resistance to prior TRK kinase inhibition in patients with TRK fusion-positive solid tumors. Cancer Discovery. 2017;7:963–72.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  13. Hyman DM, Smyth LM, Donoghue MTA, et al. AKT inhibition in solid tumors with AKT1 mutations. J Clin Oncol. 2017;35:2251–9.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Hyman DM, Piha-Paul SA, Won H, et al. HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature. 2018;554:189–94.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  16. Hechtman JF, Benayed R, Hyman DM, et al. Pan-Trk immunohistochemistry is an efficient and reliable screen for the detection of NTRK fusions. Am J Surg Pathol. 2017;41:1547–51.

    Article  PubMed  Google Scholar 

  17. Rudzinski ER, Lockwood CM, Stohr BA, et al. Pan-Trk immunohistochemistry identifies NTRK rearrangements in pediatric mesenchymal tumors. Am J Surg Pathol. 2018;

  18. Roberts KG, Li Y, Payne-Turner D, et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med. 2014;371:1005–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Braghiroli MI, Nash GM, Morris M, et al. Genomic profiling and efficacy of anti-EGFR therapy in appendiceal adenocarcinoma. J Clin Oncol. 2016;34(Suppl 4S):abstr 574.

    Article  Google Scholar 

  20. Jones DT, Hutter B, Jager N, et al. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat Genet. 2013;45:927–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Stransky N, Cerami E, Schalm S, Kim JL, Lengauer C. The landscape of kinase fusions in cancer. Nat Commun. 2014;5:4846.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Ross JS, Wang K, Gay L, et al. New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. Oncologist. 2014;19:235–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Creancier L, Vandenberghe I, Gomes B, et al. Chromosomal rearrangements involving the NTRK1 gene in colorectal carcinoma. Cancer Lett. 2015;365:107–11.

    Article  PubMed  CAS  Google Scholar 

  24. Ardini E, Bosotti R, Borgia AL, et al. The TPM3-NTRK1 rearrangement is a recurring event in colorectal carcinoma and is associated with tumor sensitivity to TRKA kinase inhibition. Mol Oncol. 2014;8:1495–507.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Shi E, Chmielecki J, Tang CM, et al. FGFR1 and NTRK3 actionable alterations in “wild-type” gastrointestinal stromal tumors. J Transl Med. 2016;14:339.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Brenca M, Rossi S, Polano M, et al. Transcriptome sequencing identifies ETV6-NTRK3 as a gene fusion involved in GIST. J Pathol. 2016;238:543–9.

    Article  PubMed  CAS  Google Scholar 

  27. Frattini V, Trifonov V, Chan JM, et al. The integrated landscape of driver genomic alterations in glioblastoma. Nat Genet. 2013;45:1141–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Zheng Z, Liebers M, Zhelyazkova B, et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med. 2014;20:1479–84.

    Article  PubMed  CAS  Google Scholar 

  29. Wu G, Diaz AK, Paugh BS, et al. The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma. Nat Genet. 2014;46:444–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Rubin BP, Chen CJ, Morgan TW, et al. Congenital mesoblastic nephroma t(12;15) is associated with ETV6-NTRK3 gene fusion: cytogenetic and molecular relationship to congenital (infantile) fibrosarcoma. Am J Pathol. 1998;153:1451–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Knezevich SR, Garnett MJ, Pysher TJ, Beckwith JB, Grundy PE, Sorensen PH. ETV6-NTRK3 gene fusions and trisomy 11 establish a histogenetic link between mesoblastic nephroma and congenital fibrosarcoma. Cancer Res. 1998;58:5046–8.

    PubMed  CAS  Google Scholar 

  32. El Demellawy D, Cundiff CA, Nasr A, et al. Congenital mesoblastic nephroma: a study of 19 cases using immunohistochemistry and ETV6-NTRK3 fusion gene rearrangement. Pathology. 2016;48:47–50.

    Article  PubMed  CAS  Google Scholar 

  33. Bourgeois JM, Knezevich SR, Mathers JA, Sorensen PH. Molecular detection of the ETV6-NTRK3 gene fusion differentiates congenital fibrosarcoma from other childhood spindle cell tumors. Am J Surg Pathol. 2000;24:937–46.

    Article  PubMed  CAS  Google Scholar 

  34. Vaishnavi A, Capelletti M, Le AT, et al. Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat Med. 2013;19:1469–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Tognon C, Knezevich SR, Huntsman D, et al. Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma. Cancer Cell. 2002;2:367–76.

    Article  PubMed  CAS  Google Scholar 

  36. Krings G, Joseph NM, Bean GR, et al. Genomic profiling of breast secretory carcinomas reveals distinct genetics from other breast cancers and similarity to mammary analog secretory carcinomas. Mod Pathol. 2017;30:1086–99.

    Article  PubMed  CAS  Google Scholar 

  37. Skalova A, Vanecek T, Sima R, et al. Mammary analogue secretory carcinoma of salivary glands, containing the ETV6-NTRK3 fusion gene: a hitherto undescribed salivary gland tumor entity. Am J Surg Pathol. 2010;34:599–608.

    PubMed  Google Scholar 

  38. Bishop JA, Yonescu R, Batista D, Begum S, Eisele DW, Westra WH. Utility of mammaglobin immunohistochemistry as a proxy marker for the ETV6-NTRK3 translocation in the diagnosis of salivary mammary analogue secretory carcinoma. Hum Pathol. 2013;44:1982–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Skalova A, Vanecek T, Simpson RH, et al. Mammary analogue secretory carcinoma of salivary glands: molecular analysis of 25 ETV6 gene rearranged tumors with lack of detection of classical ETV6-NTRK3 fusion transcript by standard RT-PCR: report of 4 cases harboring ETV6-X gene fusion. Am J Surg Pathol. 2016;40:3–13.

    Article  PubMed  Google Scholar 

  40. Wiesner T, He J, Yelensky R, et al. Kinase fusions are frequent in Spitz tumours and spitzoid melanomas. Nat Commun. 2014;5:3116.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Chiang S, Cotzia P, Hyman DM, et al. NTRK fusions define a novel uterine sarcoma subtype with features of fibrosarcoma. Am J Surg Pathol. 2018;42:791–8.

    Article  PubMed  Google Scholar 

  42. Musholt TJ, Musholt PB, Khaladj N, Schulz D, Scheumann GF, Klempnauer J. Prognostic significance of RET and NTRK1 rearrangements in sporadic papillary thyroid carcinoma. Surgery. 2000;128:984–93.

    Article  PubMed  CAS  Google Scholar 

  43. Wajjwalku W, Nakamura S, Hasegawa Y, et al. Low frequency of rearrangements of the ret and trk proto-oncogenes in Japanese thyroid papillary carcinomas. Jpn J Cancer Res. 1992;83:671–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Prasad ML, Vyas M, Horne MJ, et al. NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer. 2016;122:1097–107.

    Article  PubMed  CAS  Google Scholar 

  45. Ricarte-Filho JC, Li S, Garcia-Rendueles ME, et al. Identification of kinase fusion oncogenes in post-Chernobyl radiation-induced thyroid cancers. J Clin Invest. 2013;123:4935–44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Leeman-Neill RJ, Kelly LM, Liu P, et al. ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer. 2014;120:799–807.

    Article  PubMed  CAS  Google Scholar 

Download references

Funding

This work was supported by grants from the NIH/NCI (R01CA193837, YC; R01CA208100, YC; P50CA092629, YC; U54CA224079, YC; P50CA140146, PC; DP2 CA174499, PC; R01CA228216, PC), FDA/OPD (R01FD005731, PC), US DOD (W81XWH-10-1-0197, PC), Prostate Cancer Foundation (YC), and Bloomberg Family Foundation (NTAP Collins Scholar, PC).

Availability of data and materials

All supporting data and materials have been included within the article.

Author information

Authors and Affiliations

Authors

Contributions

Both authors wrote, revised, and approved the final manuscript.

Corresponding author

Correspondence to Ping Chi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

Both authors declare that they have no competing interests.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Y., Chi, P. Basket trial of TRK inhibitors demonstrates efficacy in TRK fusion-positive cancers. J Hematol Oncol 11, 78 (2018). https://doi.org/10.1186/s13045-018-0622-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13045-018-0622-4

Keywords