Response to Selpercatinib in a Patient With Recurrent Glioblastoma and RET Amplification

Authors:
Cameron Czech Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, North Carolina;
Department of Practice Advancement and Clinical Education, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina;
Now with Exelixis Inc., Alameda, California;

Search for other papers by Cameron Czech in
Current site
Google Scholar
PubMed
Close
 PharmD
,
Ashley Chen Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, North Carolina;
Department of Practice Advancement and Clinical Education, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina;
Now with Department of Pharmacy, University of Washington Medical Center, Seattle, Washington;

Search for other papers by Ashley Chen in
Current site
Google Scholar
PubMed
Close
 PharmD
,
Katherine P. Morgan Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, North Carolina;
Department of Practice Advancement and Clinical Education, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina;

Search for other papers by Katherine P. Morgan in
Current site
Google Scholar
PubMed
Close
 PharmD, BCOP, CPP
,
Carlos Zamora Division of Neuroradiology, Department of Radiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina;

Search for other papers by Carlos Zamora in
Current site
Google Scholar
PubMed
Close
 MD, PhD
,
Sherif El-Refai Tempus Laboratories Inc., Chicago, Illinois;

Search for other papers by Sherif El-Refai in
Current site
Google Scholar
PubMed
Close
 PharmD, PhD, MBA
,
Norleena Poynter Department of Radiation Oncology, Duke University Health System, Scotland Cancer Treatment Center, Durham, North Carolina;

Search for other papers by Norleena Poynter in
Current site
Google Scholar
PubMed
Close
 MD
, and
Simon Khagi Division of Medical Oncology, Department of Medicine, and
Department of Neurosurgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina;
Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina; and
Now with Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire.

Search for other papers by Simon Khagi in
Current site
Google Scholar
PubMed
Close
 MD
Full access

Glioblastoma (GBM) is a malignant central nervous system neoplasm that remains largely incurable. Limited treatment options currently exist after disease progression on standard-of-care first-line therapy. However, repurposing the use of approved therapies in patients with potentially targetable genomic alterations continues to be an emerging area of interest. This report presents the first description of a patient with isocitrate dehydrogenase wild-type GBM with an underlying RET amplification who demonstrated a near-complete response while receiving therapy with the RET inhibitor selpercatinib. The case highlights the excellent blood-brain barrier penetration of selpercatinib, as well as its potential role in the management of RET-amplified GBM. Larger biomarker-enriched studies are needed to confirm the results of this case report. Given the rare incidence of RET alterations in GBM, findings from this report can help guide and support optimal treatment strategies for patients with RET-altered GBM.

Background

Glioblastoma (GBM) is an aggressive central nervous system (CNS) neoplasm that remains largely incurable. Patients diagnosed with this disease have a poor prognosis, with survival rates of 14 to 15 months after diagnosis.1 Surgery, if feasible, followed by radiation therapy in combination with temozolomide is the standard of care for most patients with newly diagnosed GBM.2,3 Additionally, repurposing the use of approved targeted therapies in patients with specific genomic alterations or those with various genomic or metabolic abnormalities continues to be an emerging area of interest.4 The selective RET inhibitor selpercatinib is poised to alter the treatment landscape for patients with RET-altered malignancies. Currently, selpercatinib has FDA-approved indications for the treatment of metastatic RET fusion–positive non–small cell lung cancer (NSCLC) and thyroid cancers, as well as RET-mutant medullary thyroid cancer (MTC) requiring systemic treatment.5 Recent evidence has also demonstrated antitumor activity in additional RET fusion–driven advanced solid tumors.6 However, the implications of genomic alterations outside of RET fusion–positive and RET mutation–positive disease have not been fully characterized. Thus, the safety and efficacy of selpercatinib has not been fully described in solid tumors with additional RET alterations, specifically those harboring RET amplification.

This report presents the first description of a patient with recurrent GBM and a concurrent RET amplification who demonstrated clinically significant disease response while receiving selpercatinib therapy. The patient’s consent was obtained for publication of this manuscript.

Case Presentation

A 48-year-old man with no significant past medical history presented with sudden onset of severe right-sided headache and emesis. MRI revealed 2 rim-enhancing lesions in the right temporal lobe. The patient underwent a gross total resection of the lesions, with surgical pathology revealing a GBM. MGMT promoter was unmethylated, and no IDH mutations were detected. The tumor sample was clinically profiled at a College of American Pathologists–accredited, CLIA-certified laboratory (Tempus Laboratories) using the Tempus xT laboratory developed test (LDT), similar to previously described methods.7 Additional review of next-generation sequencing (NGS) revealed ATM exon 10 splice acceptor mutation, CDK4 amplification, MDM2 amplification, and RET amplification.

The patient underwent radiation therapy with concurrent temozolomide. The presence of an ATM mutation led to the initiation of olaparib with adjuvant temozolomide following completion of chemoradiotherapy. Additionally, he was treated with an externally applied, alternating tumor-treating field treatment (Optune, Novocure).8 Four cycles of maintenance temozolomide + olaparib therapy were completed but were subsequently discontinued due to disease progression. Pseudoprogression was a consideration; however, the patient met RANO criteria given his MRI findings, proximity to completing chemoradiotherapy (>12 weeks), ongoing need for steroids, and worsening neurologic symptoms.9 As mentioned previously, further evaluation of genomic sequencing demonstrated the presence of a RET amplification. These findings, in addition to the recently published efficacy data regarding the use of selpercatinib in patients with metastatic NSCLC to the brain with known RET-mutated disease, provided rationale for the use of selpercatinib upon disease progression.10

Selpercatinib was dosed at 160 mg twice daily as a continuous regimen. After 6 weeks of selpercatinib treatment, MRI results demonstrated near-complete resolution of gadolinium-enhanced disease on MRI, with a significant reduction in mass effect and vasogenic edema (Figure 1). Seizure activity had also resolved. Repeat MRI results after 12 weeks of therapy continued to show a positive response to therapy with no new areas of disease. After 12 weeks of treatment, selpercatinib was held due to treatment-related liver enzyme elevation. After liver enzyme normalization, selpercatinib was restarted and titrated back to full-dose therapy. During this period of drug titration, imaging revealed a partial response. However, after 8 months of selpercatinib treatment, repeat MRI revealed unequivocal disease progression. Selpercatinib was discontinued and the patient was initiated on the next line of therapy.

Figure 1.
Figure 1.

MRI examinations 8 weeks prior to progression (A, B, C), at the time of progression (D, E, F), and 6 weeks after initiation of selpercatinib (G, H, I). Postcontrast T1 at the time of progression (D) shows a significant increase in heterogeneous enhancement in the right temporal lobe with a discrete nodule along the posterior aspect of the lesion (arrows). Corresponding FLAIR image (E) shows markedly increased edema and associated mass effect compared with 8 weeks prior (B). Diffusion-weighted image (F) shows a new area of restricted diffusion posteriorly (arrowheads). MRI 6 weeks after initiating selpercatinib shows near complete resolution of contrast enhancement (G), significant reduction in mass effect and edema (H), and resolution of restricted diffusion (I).

Citation: Journal of the National Comprehensive Cancer Network 20, 9; 10.6004/jnccn.2022.7030

Discussion

Progression Versus Pseudoprogression

Although our patient met standard RANO criteria for disease progression, we thought pseudoprogression was possible given exposure to olaparib while on maintenance temozolomide. As reported by Baxter et al,11 the rate of pseudoprogression approached 20% in a cohort of children exposed to the combination of veliparib and radiation, followed by maintenance temozolomide. However, it is worth mentioning that a meta-analysis of 73 studies in high-grade glioma by Abbasi et al12 reported a 36% incidence of pseudoprogression in patients treated with standard therapy. Therefore, we speculate that the presence of a PARP inhibitor did not appear to add to the risk of pseudoprogression, considering that the likelihood of pseudoprogression is seemingly higher with standard-of-care therapy. From a molecular standpoint, MGMT promotor unmethylated disease is more likely to progress early during treatment.13 The possibility of pseudoprogression was further diminished given that the patient did not improve on increasing doses of steroids.

With our patient starting selpercatinib, followed by his rapid clinical and radiographic improvement, it was favored that this was true progression of disease. However, it is also plausible that selpercatinib, although being a highly specific RET inhibitor, could induce a response similar to bevacizumab given that RET signaling may be involved in crosstalk with the VEGF pathway.14 This is further supported by selpercatinib having the known adverse effects of hypertension and impaired wound healing.5

RET-Altered Disease and Current Practices

Considered a proto-oncogene, the RET gene encodes a transmembrane receptor tyrosine kinase that is composed of a large extracellular domain, an intracellular kinase, and a transmembrane domain.15 Alterations in RET are primarily activated through chromosomal rearrangements leading to gene fusions or through specific point mutations. Activation of oncogenicity through RET-fusion proteins leads to ligand-independent dimerization, constitutively active RET, and ultimately uncontrolled cellular proliferation.10 Multikinase inhibitors (MKIs) with RET inhibitor activity, such as cabozantinib and vandetanib, have historically been used for the targeted treatment of RET-driven cancers.16,17 However, both agents are associated with limited overall and duration of responses, as well as risk of a variety of off-target adverse effects.1619

Advances in the field of RET-targeted therapies have led to the recent approval of selpercatinib. Selpercatinib is a novel, ATP-competitive, highly selective small-molecule RET kinase inhibitor with significant CNS penetration and low potential for drug interactions (Figure 2). This agent has demonstrated an improved efficacy and safety profile compared with previous agents used to target malignancies with known RET-fusion alterations. Currently, selpercatinib is approved for the treatment of adult patients with metastatic RET fusion–positive NSCLC and thyroid cancer, as well as adult and pediatric patients with RET-mutant MTC requiring systemic therapy.5 Selpercatinib has demonstrated response rates of 64% to 85% in patients previously treated with a RET targeted therapy and those who are treatment-naïve.10,20 Selpercatinib demonstrated lasting and durable clinical responses in those who respond to therapy yielding significant progression-free survival rates and durations of response. Additionally, selpercatinib has demonstrated clinical activity among patients with known CNS metastases. Of the 11 patients with NSCLC previously treated with chemotherapy who had measurable CNS disease, 91% had a clinical response (3 complete responses, 7 partial responses, and 1 stable disease), with a median CNS duration of response of 10.1 months.10

Figure 2.
Figure 2.

Selpercatinib is a highly selective, ATP-competitive, RET receptor tyrosine kinase inhibitor. Binding of selpercatinib to RET-fusion or point-mutated RET receptors results in inhibition of RET kinase activity and changes in intracellular signaling pathways which lead to decreased cellular proliferation and increased cellular apoptosis.

Reproduced with permission from Eli Lilly and Company.

Citation: Journal of the National Comprehensive Cancer Network 20, 9; 10.6004/jnccn.2022.7030

Pharmacokinetics and Pharmacodynamics of Selpercatinib and Blood–Brain Barrier Penetration

For drugs used in the treatment of CNS-related diseases, attaining the targetable concentrations needed for a clinical response is complicated by the presence of the blood–brain barrier (BBB). The BBB plays an essential role in maintaining homeostasis within the CNS environment and is equipped with a wide array of both influx and efflux transport proteins that regulate concentration ion gradients, the delivery of macromolecules to meet the energy needs of the brain, and prevents the entry of numerous, potentially harmful substances from entering the CNS environment.2123 Evaluation of currently available literature has demonstrated varying levels of CNS penetration of tyrosine kinase inhibitors (TKIs) with activity against RET (Table 1).2431 The CNS penetration of selpercatinib has been demonstrated in both in vitro and in vivo mouse tumor models and multiple patient case reports.32,33 As described previously, the ability of selpercatinib to effectively cross the BBB has been further supported by the efficacy and responses demonstrated in patients with RET-altered NSCLC who had confirmed metastases to the brain.10

Table 1.

Comparison of TKIs With Activity Against RET

Table 1.

The dose–response relationship of selpercatinib has not been fully characterized. However, depending on the RET alteration, the IC50 for selpercatinib ranges from 0.92 to 67.8 nanomolar.5 Given the high selectivity and potency of selpercatinib for multiple RET alterations, limited concentrations of active drug are needed to permeate the BBB to have an effect at the site of action if present within the CNS space. Although selpercatinib has the highest specificity for inhibition of RET alterations, achievement of higher concentrations leads to inhibition of additional tyrosine kinases, including various isoforms of VEGF and fibroblast growth factor receptor, both of which have isoforms that have been implicated in the growth and survival of various CNS malignancies.5,15,34 These characteristics provide a better reasoning and rationale for the effective CNS penetration seen with selpercatinib and its potential use in patients with known RET-altered GBM.

Targeting RET Alterations and Potential Off-Target Activities

The complexity of genomic changes seen in RET-driven malignancies highlights the need for further investigation of this mutation. RET rearrangements are mutually exclusive with other driver mutations.35 In NSCLC, at least 12 different gene partners have been described for RET mutations, including KIF5B8, CCDC6, MYO5C, TRIM33, and ERC1.35,36 Functional studies have shown that inhibitors of RET are capable of inhibiting a wide range of RET alterations, including gatekeeper mutations V804L and V804M, KIF5B-RE, CCDC6-RET, M918T, and C634W.34 As their name suggests, MKIs target a spectrum of kinases outside of RET. Due to the similarities in homology between the kinase domain of RET and other TKIs, MKIs may have activity against RET mutations at higher concentrations. However, the off-target effects of MKIs can lead to inferior inhibition of RET and increased incidence of adverse events, leading to potential dose modifications or drug discontinuation. VEGFR2 inhibitors, such as cabozantinib and vandetanib, have different efficacies against various RET mutations and can effectively block the activity of RET M918T. However, they are unable to inhibit gatekeeper mutations, including RET V804M and RET V804L, and may lead to mechanisms of acquired resistance to various MKIs.15,35

According to the AACR Project GENIE Consortium, RET is altered in approximately 2.75% of malignant CNS neoplasms, with a total of 37 glioblastoma and 15 anaplastic astrocytoma cases reported.37 A similar prevalence of RET-altered glioma cases have been reported in the cBioPortal for Cancer Genomics, with an incidence of 6.6%.38,39 Overexpression of RET kinase has only been identified in 7 glioma tissue samples per the Catalogue of Somatic Mutations in Cancer (COSMIC) database.40 Woo et al41 conducted NGS in 356 diffuse gliomas and identified 2 patients harboring oncogenic RET-fusion mutations. With such a low prevalence, potential therapeutic targets for RET-mutated GBM have not been evaluated in either the preclinical or clinical stages of drug development.

Response to selpercatinib in the setting of RET amplification has not been previously reported. To better understand potential mechanisms surrounding the observed response in this case report, further analysis of the tumor sample was performed using whole-exome sequencing. A low-level ANKRD26-RET fusion was detected in the DNA, with 12 supporting reads. NGS analysis confirmed that this fusion was not detected in the RNA, even with low support. Additionally, the RET kinase domain was retained, signifying that this would be predicted to be oncogenic in nature. However, due to the ANKRD26-RET fusion low level of support in the DNA and absence within the RNA, it is difficult to determine whether this was a true RET-fusion or rather sequencing noise that may have arisen as a side effect of the RET amplification. Although limited, low-level fusion events in the setting of specific gene copy number amplifications have been previously described in patients with newly diagnosed GBM via the proposed breakage-fusion-bridge cycle model, specifically in the setting of EGFR copy number amplification.42 Given the lack of fusion protein detected in the tumor RNA, it can be theorized that a fusion protein cannot be ultimately transcribed and would not be a factor in the process of tumorigenesis. This suggests that selpercatinib could have activity against the RET kinase domain and that RET amplification revealed in this case could be the primary driver of the observed response.

Conclusions

Some of the primary challenges associated with the treatment of GBM include the inability of many systemic therapies to effectively penetrate the BBB and the heterogeneity of oncogenic drivers at the molecular level. As described in this report, selpercatinib provided a complete and durable clinical response in this patient with RET-amplified GBM who, as we highly suspect, was experiencing true disease progression. Activity against RET-amplified disease has not been previously described with selpercatinib. However, this report provides clinical evidence that selpercatinib has high specificity and activity against RET tyrosine kinases that are overexpressed and acting as an underlying oncogenic driver. Additionally, this report further supports the ability of selpercatinib to effectively cross the BBB in concentrations high enough to produce a profound clinical response.

To our knowledge, this is the first case report that describes a patient with RET-amplified disease being treated with and responding to selpercatinib therapy. Given the rare occurrence of RET alterations in GBM, our experience with this case may help inform future biomarker-enriched studies within this population.

Acknowledgments

We thank Jay White, Pablo Lee, and Boris Kin Lim from Eli Lilly and Company for providing editorial assistance under the guidance of the authors and allowing us to use the image detailing selpercatinib’s mechanism of action for the purposes of this case report. We thank Carlos Zamora for providing excellent review and analysis of the MRI imaging surrounding the clinical response described in this case report. Additionally, we thank Tempus Laboratory Inc. for contributing key insights into the genomic analysis detailed in this report.

References

  • 1.

    Hanif F, Muzaffar K, Perveen K, et al. Glioblastoma multiforme: a review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pac J Cancer Prev 2017;18:39.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 2.

    Le Rhun E, Preusser M, Roth P, et al. Molecular targeted therapy of glioblastoma [published online September 11, 2019]. Cancer Treat Rev, doi:10.1016/j,ctrv.2019.101896

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Nabors LB, Portnow J, Baehring J, et al. NCCN Clinical Practice Guidelines in Oncology: Central Nervous System Cancers. Version 2.2021. Accessed June 15, 2021. To view the most recent version, visit NCCN.org

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Kurzrock R. Selpercatinib aimed at RET-altered cancers. N Engl J Med 2020;383:868869.

  • 5.

    Retevmo (selpercatinib) [package insert]. Indianapolis, IN: Eli Lilly and Company; 2020.

  • 6.

    Bauer TM, Besse B, Loong HHF, et al. Safety of selpercatinib for RET-altered advanced solid tumors: a post hoc analysis of LIBRETTO-001 [abstract]. Presented at the 2021 Annual Meeting of the American Association for Cancer Research; April 10–15, 2021. Abstract CT160.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7.

    Beaubier N, Tell R, Lau D, et al. Clinical validation of the tempus xT next-generation targeted oncology sequencing assay. Oncotarget 2019;10:23842396.

  • 8.

    Stupp R, Taillibert S, Kanner A, et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. JAMA 2017;318:23062316.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 2010;28:19631972.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Drilon A, Oxnard GR, Tan DSW, et al. Efficacy of selpercatinib in RET fusion–positive non–small-cell lung cancer. N Engl J Med 2020;383:813824.

  • 11.

    Baxter PA, Su JM, Onar-Thomas A, et al. A phase I/II study of veliparib (ABT-888) with radiation and temozolomide in newly diagnosed diffuse pontine glioma: a Pediatric Brain Tumor Consortium study. Neuro Oncol 2020;22:875885.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Abbasi AW, Westerlaan HE, Holtman GA, et al. Incidence of tumor progression and pseudoprogression in high-grade gliomas: a systematic review and meta-analysis. Clin Neuroradiol 2018;28:401411.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Brandes AA, Franceschi E, Tosoni A, et al. MGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol 2008;26:21922197.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Tufro A, Teichman J, Banu N, et al. Crosstalk between VEGF-A/VEGFR2 and GDNF/RET signaling pathways. Biochem Biophys Res Commun 2007;358:410416.

  • 15.

    Subbiah V, Yang D, Velcheti V, et al. State-of-the-art strategies for targeting RET-dependent cancers. J Clin Oncol 2020;38:12091221.

  • 16.

    Caprelsa (vandetanib) [package insert]. Wilmington, DE: AstraZeneca Pharmaceuticals; 2011

  • 17.

    Cabometyx (cabozantinib) [package insert]. Alameda, CA: Exelixis, Inc; 2012

  • 18.

    Wells SA Jr, Robinson BG, Gagel RF, et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. J Clin Oncol 2012;30:134141.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Schlumberger M, Elisei R, Müller S, et al. Overall survival analysis of EXAM, a phase III trial of cabozantinib in patients with radiographically progressive medullary thyroid carcinoma. Ann Oncol 2017;28:28132819.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Wirth LJ, Sherman E, Robinson B, et al. Efficacy of selpercatinib in RET-altered thyroid cancers. N Engl J Med 2020;383:825835.

  • 21.

    Pajouhesh H, Lenz GR. Medicinal chemical properties of successful central nervous system drugs. NeuroRx 2005;2:541553.

  • 22.

    On NH, Yathindranath V, Sun Z, et al. Pathways for drug delivery to the central nervous system. In: Wang B, Hu L, Siahaan TJ, eds. Drug Delivery: Principles and Applications. 2nd ed. Hoboken, NJ: John Wiley & Sons, Inc; 2016:353382.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Saunders NR, Habgood MD, Møllgård K, et al. The biological significance of brain barrier mechanisms: help or hindrance in drug delivery to the central nervous system? F1000Res 2016;5:313.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Cloughesy TF, Drappatz J, de Groot J, et al. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients with prior antiangiogenic therapy. Neuro Oncol 2018;20:259267.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Wen PY, Drappatz J, de Groot J, et al. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients naive to antiangiogenic therapy. Neuro Oncol 2018;20:249258.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Gainor JF. Clinical activity and tolerability of BLU-667, a highly potent and selective RET inhibitor, in patients with advanced RET-fusion+ non-small cell lung cancer Presented at the 2019 ASCO Annual Meeting; May 31–June 4, 2019; Chicago, IL.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Kreisl TN, McNeill KA, Sul J, et al. A phase I/II trial of vandetanib for patients with recurrent malignant glioma. Neuro Oncol 2012;14:15191526.

  • 28.

    Grisanti S, Ferrari VD, Buglione M, et al. Second line treatment of recurrent glioblastoma with sunitinib: results of a phase II study and systematic review of literature. J Neurosurg Sci 2019;63:458467.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Nabors LB, Supko JG, Rosenfeld M, et al. Phase I trial of sorafenib in patients with recurrent or progressive malignant glioma. Neuro Oncol 2011;13:13241330.

  • 30.

    Norden AD, Schiff D, Ahluwalia MS, et al. Phase II trial of triple tyrosine kinase receptor inhibitor nintedanib in recurrent high-grade gliomas. J Neurooncol 2015;121:297302.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Muhic A, Poulsen HS, Sorensen M, et al. Phase II open-label study of nintedanib in patients with recurrent glioblastoma multiforme. J Neurooncol 2013;111:205212.

  • 32.

    Drilon A, Lin JJ, Filleron T, et al. Frequency of brain metastases and multikinase inhibitor outcomes in patients with RET–rearranged lung cancers. J Thorac Oncol 2018;13:15951601.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Subbiah V, Velcheti V, Tuch BB, et al. Selective RET kinase inhibition for patients with RET-altered cancers. Ann Oncol 2018;29:18691876.

  • 34.

    Jimenez-Pascual A, Siebzehnrubl FA. Fibroblast growth factor receptor functions in glioblastoma. Cells 2019;8:715.

  • 35.

    Drilon A, Fu S, Patel MR, et al. A phase I/Ib trial of the VEGFR-sparing multikinase RET inhibitor RXDX-105. Cancer Discov 2019;9:384395.

  • 36.

    Reardon DA, Turner S, Peters KB, et al. A review of VEGF/VEGFR-targeted therapeutics for recurrent glioblastoma. J Natl Compr Canc Netw 2011;9:414427.

  • 37.

    AACR Project GENIE Consortium. AACR project GENIE: powering precision medicine through an international consortium. Cancer Discov 2017;7:818831.

  • 38.

    Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401404.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39.

    Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1.

  • 40.

    Sanger Institute. COSMIC - Catalogue of Somatic Mutations in Cancer. Accessed May 24, 2021. Available at: http://cancer.sanger.ac.uk/cosmic

  • 41.

    Woo HY, Na K, Yoo J, et al. Glioblastomas harboring gene fusions detected by next-generation sequencing. Brain Tumor Pathol 2020;37:136144.

  • 42.

    Lopez-Gines C, Gil-Benso R, Ferrer-Luna R, et al. New pattern of EGFR amplification in glioblastoma and the relationship of gene copy number with gene expression profile. Mod Pathol 2010;23:856865.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation

Submitted August 4, 2021; final revision received May 8, 2022; accepted for publication May 9, 2022.

Disclosures: Dr. Czech has disclosed being employed by Exelixis, Inc. Dr. El-Refai has disclosed owning stock and having other ownership interests in Tempus Labs, Inc. The remaining authors have disclosed that they have not received any financial consideration from any person or organization to support the preparation, analysis, results, or discussion of this article.

Correspondence: Simon Khagi, MD, Dartmouth Cancer Center Manchester, Notre Dame Pavilion, 87 McGregor Street, Manchester, NH 03102. Email: simon.khagi@hitchcock.org
  • Collapse
  • Expand
  • Figure 1.

    MRI examinations 8 weeks prior to progression (A, B, C), at the time of progression (D, E, F), and 6 weeks after initiation of selpercatinib (G, H, I). Postcontrast T1 at the time of progression (D) shows a significant increase in heterogeneous enhancement in the right temporal lobe with a discrete nodule along the posterior aspect of the lesion (arrows). Corresponding FLAIR image (E) shows markedly increased edema and associated mass effect compared with 8 weeks prior (B). Diffusion-weighted image (F) shows a new area of restricted diffusion posteriorly (arrowheads). MRI 6 weeks after initiating selpercatinib shows near complete resolution of contrast enhancement (G), significant reduction in mass effect and edema (H), and resolution of restricted diffusion (I).

  • Figure 2.

    Selpercatinib is a highly selective, ATP-competitive, RET receptor tyrosine kinase inhibitor. Binding of selpercatinib to RET-fusion or point-mutated RET receptors results in inhibition of RET kinase activity and changes in intracellular signaling pathways which lead to decreased cellular proliferation and increased cellular apoptosis.

    Reproduced with permission from Eli Lilly and Company.

  • 1.

    Hanif F, Muzaffar K, Perveen K, et al. Glioblastoma multiforme: a review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pac J Cancer Prev 2017;18:39.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 2.

    Le Rhun E, Preusser M, Roth P, et al. Molecular targeted therapy of glioblastoma [published online September 11, 2019]. Cancer Treat Rev, doi:10.1016/j,ctrv.2019.101896

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Nabors LB, Portnow J, Baehring J, et al. NCCN Clinical Practice Guidelines in Oncology: Central Nervous System Cancers. Version 2.2021. Accessed June 15, 2021. To view the most recent version, visit NCCN.org

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Kurzrock R. Selpercatinib aimed at RET-altered cancers. N Engl J Med 2020;383:868869.

  • 5.

    Retevmo (selpercatinib) [package insert]. Indianapolis, IN: Eli Lilly and Company; 2020.

  • 6.

    Bauer TM, Besse B, Loong HHF, et al. Safety of selpercatinib for RET-altered advanced solid tumors: a post hoc analysis of LIBRETTO-001 [abstract]. Presented at the 2021 Annual Meeting of the American Association for Cancer Research; April 10–15, 2021. Abstract CT160.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7.

    Beaubier N, Tell R, Lau D, et al. Clinical validation of the tempus xT next-generation targeted oncology sequencing assay. Oncotarget 2019;10:23842396.

  • 8.

    Stupp R, Taillibert S, Kanner A, et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. JAMA 2017;318:23062316.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 2010;28:19631972.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Drilon A, Oxnard GR, Tan DSW, et al. Efficacy of selpercatinib in RET fusion–positive non–small-cell lung cancer. N Engl J Med 2020;383:813824.

  • 11.

    Baxter PA, Su JM, Onar-Thomas A, et al. A phase I/II study of veliparib (ABT-888) with radiation and temozolomide in newly diagnosed diffuse pontine glioma: a Pediatric Brain Tumor Consortium study. Neuro Oncol 2020;22:875885.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Abbasi AW, Westerlaan HE, Holtman GA, et al. Incidence of tumor progression and pseudoprogression in high-grade gliomas: a systematic review and meta-analysis. Clin Neuroradiol 2018;28:401411.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Brandes AA, Franceschi E, Tosoni A, et al. MGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol 2008;26:21922197.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Tufro A, Teichman J, Banu N, et al. Crosstalk between VEGF-A/VEGFR2 and GDNF/RET signaling pathways. Biochem Biophys Res Commun 2007;358:410416.

  • 15.

    Subbiah V, Yang D, Velcheti V, et al. State-of-the-art strategies for targeting RET-dependent cancers. J Clin Oncol 2020;38:12091221.

  • 16.

    Caprelsa (vandetanib) [package insert]. Wilmington, DE: AstraZeneca Pharmaceuticals; 2011

  • 17.

    Cabometyx (cabozantinib) [package insert]. Alameda, CA: Exelixis, Inc; 2012

  • 18.

    Wells SA Jr, Robinson BG, Gagel RF, et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. J Clin Oncol 2012;30:134141.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Schlumberger M, Elisei R, Müller S, et al. Overall survival analysis of EXAM, a phase III trial of cabozantinib in patients with radiographically progressive medullary thyroid carcinoma. Ann Oncol 2017;28:28132819.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Wirth LJ, Sherman E, Robinson B, et al. Efficacy of selpercatinib in RET-altered thyroid cancers. N Engl J Med 2020;383:825835.

  • 21.

    Pajouhesh H, Lenz GR. Medicinal chemical properties of successful central nervous system drugs. NeuroRx 2005;2:541553.

  • 22.

    On NH, Yathindranath V, Sun Z, et al. Pathways for drug delivery to the central nervous system. In: Wang B, Hu L, Siahaan TJ, eds. Drug Delivery: Principles and Applications. 2nd ed. Hoboken, NJ: John Wiley & Sons, Inc; 2016:353382.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Saunders NR, Habgood MD, Møllgård K, et al. The biological significance of brain barrier mechanisms: help or hindrance in drug delivery to the central nervous system? F1000Res 2016;5:313.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Cloughesy TF, Drappatz J, de Groot J, et al. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients with prior antiangiogenic therapy. Neuro Oncol 2018;20:259267.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Wen PY, Drappatz J, de Groot J, et al. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients naive to antiangiogenic therapy. Neuro Oncol 2018;20:249258.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Gainor JF. Clinical activity and tolerability of BLU-667, a highly potent and selective RET inhibitor, in patients with advanced RET-fusion+ non-small cell lung cancer Presented at the 2019 ASCO Annual Meeting; May 31–June 4, 2019; Chicago, IL.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Kreisl TN, McNeill KA, Sul J, et al. A phase I/II trial of vandetanib for patients with recurrent malignant glioma. Neuro Oncol 2012;14:15191526.

  • 28.

    Grisanti S, Ferrari VD, Buglione M, et al. Second line treatment of recurrent glioblastoma with sunitinib: results of a phase II study and systematic review of literature. J Neurosurg Sci 2019;63:458467.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Nabors LB, Supko JG, Rosenfeld M, et al. Phase I trial of sorafenib in patients with recurrent or progressive malignant glioma. Neuro Oncol 2011;13:13241330.

  • 30.

    Norden AD, Schiff D, Ahluwalia MS, et al. Phase II trial of triple tyrosine kinase receptor inhibitor nintedanib in recurrent high-grade gliomas. J Neurooncol 2015;121:297302.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Muhic A, Poulsen HS, Sorensen M, et al. Phase II open-label study of nintedanib in patients with recurrent glioblastoma multiforme. J Neurooncol 2013;111:205212.

  • 32.

    Drilon A, Lin JJ, Filleron T, et al. Frequency of brain metastases and multikinase inhibitor outcomes in patients with RET–rearranged lung cancers. J Thorac Oncol 2018;13:15951601.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Subbiah V, Velcheti V, Tuch BB, et al. Selective RET kinase inhibition for patients with RET-altered cancers. Ann Oncol 2018;29:18691876.

  • 34.

    Jimenez-Pascual A, Siebzehnrubl FA. Fibroblast growth factor receptor functions in glioblastoma. Cells 2019;8:715.

  • 35.

    Drilon A, Fu S, Patel MR, et al. A phase I/Ib trial of the VEGFR-sparing multikinase RET inhibitor RXDX-105. Cancer Discov 2019;9:384395.

  • 36.

    Reardon DA, Turner S, Peters KB, et al. A review of VEGF/VEGFR-targeted therapeutics for recurrent glioblastoma. J Natl Compr Canc Netw 2011;9:414427.

  • 37.

    AACR Project GENIE Consortium. AACR project GENIE: powering precision medicine through an international consortium. Cancer Discov 2017;7:818831.

  • 38.

    Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401404.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39.

    Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1.

  • 40.

    Sanger Institute. COSMIC - Catalogue of Somatic Mutations in Cancer. Accessed May 24, 2021. Available at: http://cancer.sanger.ac.uk/cosmic

  • 41.

    Woo HY, Na K, Yoo J, et al. Glioblastomas harboring gene fusions detected by next-generation sequencing. Brain Tumor Pathol 2020;37:136144.

  • 42.

    Lopez-Gines C, Gil-Benso R, Ferrer-Luna R, et al. New pattern of EGFR amplification in glioblastoma and the relationship of gene copy number with gene expression profile. Mod Pathol 2010;23:856865.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation

Metrics

All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 2904 522 25
PDF Downloads 1671 424 27
EPUB Downloads 0 0 0