ArticlePDF Available

Abstract

In this report, select key studies presented at the American Society of Clinical Oncology (ASCO) 2021 annual meeting are reviewed. Two major phase III randomized controlled trials were presented at the meeting: GEINO 1401 and EORTC 1709/CCTG CE.8. Both are reviewed in this report. Moreover, important phase II trials, including Alliance A0716701, and key phase I trials are included. All trials presented cover important advances in the understanding of primary brain tumor management. In addition, case series papers, trials in progress and select work on exploratory CSF biomarkers are reviewed. Altogether, research presented at ASCO 2021 highlights important advances in neuro-oncologic topics that may inform future research and practice.
Conference Report
For reprint orders, please contact: reprints@futuremedicine.com
American Society of Clinical Oncology 2021
Annual Meeting updates on primary brain
tumors and CNS metastatic tumors
Archit B Baskaran1, Priya Kumthekar2,3, Amy B Heimberger3,4 & Rimas V Lukas*,2,3
1Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
2Department of Neurology, Northwestern University, Chicago, IL, USA
3Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, IL, USA
4Department of Neurosurgery, Northwestern University, Chicago, IL, USA
*Author for correspondence: Rimas.lukas@nm.org
In this report, select key studies presented at the American Society of Clinical Oncology (ASCO) 2021 annual
meeting are reviewed. Two major phase III randomized controlled trials were presented at the meeting:
GEINO 1401 and EORTC 1709/CCTG CE.8. Both are reviewed in this report. Moreover, important phase II
trials, including Alliance A0716701, and key phase I trials are included. All trials presented cover important
advances in the understanding of primary brain tumor management. In addition, case series papers, trials
in progress and select work on exploratory CSF biomarkers are reviewed. Altogether, research presented
at ASCO 2021 highlights important advances in neuro-oncologic topics that may inform future research
and practice.
First draft submitted: 4 August 2021; Accepted for publication: 22 September 2021; Published online:
21 October 2021
Keywords: brain metastases craniopharyngioma glioblastoma leptomeningeal metastases meeting report
neuro-oncology
The American Society of Clinical Oncology (ASCO) held its annual meeting virtually 4–8 June 2021. A wide
spectrum of health professionals and researchers participated in the meeting, covering all aspects of oncology.
Highlights of the neuro-oncologic topics are reviewed here with a focus on clinical studies of primary brain tumors
and cerebrospinal fluid biomarkers in central nervous system metastases. Due to the broad scope of the meeting,
select key clinical studies will be covered in this overview. The target group of this review includes: neuro-oncologists,
neurosurgeons, radiation oncologists and general oncologists. The research included in this review was chosen due
to relevance to the target audience. This involved subjectivity on the part of the authors, and all key work was
unable to be included.
Advances in primary brain tumors: phase III trials
Two late-stage clinical trials were presented that did not meet the primary endpoints. In Grupo Espanol de
Investigacion en Neurooncologia (GEINO) 1401, the optimal number of adjuvant temozolomide cycles for
patients with newly diagnosed glioblastoma was prospectively investigated [1]. Prior retrospective meta-analyses
had suggested that there was no enhanced survival benefit for treating patients with more than six temozolomide
cycles [2]; however, this had not been determined in a prospective, randomized fashion. GEINO 1401 enrolled
159 newly diagnosed nonprogressive glioblastoma subjects who had completed standard of care radiation with
concurrent temozolomide [3]. After six adjuvant temozolomide cycles, patients were randomized 1:1 to either
observation or six additional cycles of temozolomide. Study participants were stratified by MGMT-methylation
status and by residual measurable disease. Median overall survival (OS) was not significantly different between the
two groups: 22.0 months observation versus 18.2 months additional temozolomide, HR of 0.957 (95% CI: 0.806–
1.136; p = 0.615). Two-year survival rates were also not different: 62% for the observation group versus 61% for
the additional temozolomide group. MGMT methylation favorably influenced outcomes, whereas IDH mutational
Future O ncol. (2021) 17(33), 4425–4429 ISSN 1479-6694 442510.2217/fon-2021-0955 C
2021 Future Medicine Ltd
Conference Report Baskaran, Kumthekar, Heimberger & Lukas
status and residual disease did not. This aligns with traditional dogma, which finds that patients with glioblastoma
containing a methylated MGMT promoter benefit from temozolamide [4].
The phase III trial EORTC 1709/CCTG CE.8 examined the effectiveness of the proteasome inhibitor mari-
zomib in combination with standard of care for treatment of newly diagnosed glioblastoma [5]. Patients (n = 749)
were randomized 1:1 to either radiation with concurrent temozolomide, followed by six adjuvant cycles of temo-
zolomide or with the addition of marizomib beginning at the start of radiation. Marizomib was selected based
on preclinical data and its blood–brain barrier (BBB) penetration properties [6]. No difference in median OS was
noted: 15.9 months for the control cohort versus 15.7 months for the marizomib cohort (HR: 0.99). Additionally,
there was no difference in PFS between the two arms: 6.1 months control versus 6.2 months for marizomib (HR:
1.02). Adverse events (AEs) such as ataxia, hallucinations and headache were doubled in the marizomib-treated
group.
Advances in primary brain tumors: phase II trials
The clinical trial that will likely change the standard of care for craniopharyngioma patients is Alliance A071601.
This phase II trial (n = 36) used the combination of BRAF and MEK inhibitors (vemurafenib and cobimetinib,
respectively) for subjects with the BRAF V600E mutated papillary craniopharyngioma subtype [7]. All patients
underwent prior surgical resection. BRAF mutation was demonstrated via immunohistochemistry. This study was
based on previously published work demonstrating radiographic responses targeting this pathway in this patient
population [8,9] who have a very high incidence of BRAF mutations [10]. Patients receive oral vemurafenib twice
per day for 28 days concurrently with daily oral cobimetinib for 21 days. Results from Cohort A were presented
that evaluated subjects with no prior radiation (n = 16). Fifteen patients in Cohort A had evaluable volumetric
data. Among these patients, 14 demonstrated a response as defined by at least a 20% decrease in volume. The
single nonresponder received only 2 days of treatment, which was discontinued due to toxicity. The median tumor
volume reduction in Cohort A was 83%, and median progression-free survival (PFS) was not reached at a median
follow-up of 22 months. Seventy-five percent of patients experienced grade 3 toxicity, predominantly rash. Thirteen
percent of patients developed grade 4 toxicity (hyperglycemia, elevated creatine kinase). These exceptional response
rates are the first such examples of response to systemic therapy in this patient population within the context of
a prospective trial. Cohort B results have not yet been released but uses the same approach in patients who have
progressed after radiation.
Another randomized phase II trial evaluated two dosing regimens of the anti-VEGF antibody bevacizumab
(10 mg/kg intravenously [iv.] every 2 weeks [standard] vs 3 mg/kg iv. every 2 weeks [reduced]) used in combination
with a fixed dose of the anti-PD-1 antibody nivolumab (240 mg iv. every 2 weeks) in patients with recurrent
glioblastoma [11]. Patients with recurrent glioblastoma (n = 90) at first recurrence were randomized 1:1 and
stratified by age, Karnofsky Performance Score (KPS), extent of tumor resection and MGMT methylation status.
OS at 12 months (OS12) in the overall patient population was not significantly different. Subgroup analysis in older
adult patients (>60 years old) demonstrated a better OS12 for subjects who received the standard dose bevacizumab
(46.2 vs. 23.8%) and median OS (10.6 vs 5.9 months; p = 0.046). Grade 3/4 toxicities were as anticipated. A
prior phase II study has shown that there is no therapeutic benefit of the addition of anti-VEGF therapy to PD-1
blockade in recurrent glioblastoma [12]. The current study clarifies that the dosing of the bevacizumab (high dose
vs low dose) does not influence outcome in combination with anti-PD-1.
On the basis of preclinical studies of sensitivity to PARP inhibition in IDH-mutated gliomas, a phase II clinical
trial of olaparib was evaluated in recurrent IDH-mutant high-grade glioma (OLAGLI) patients (n = 35 total;
oliogodendroglioma n = 16; astrocytoma n = 14) [13–15]. The treatment was well tolerated, but median PFS was
only 2 months. Six-month PFS (PFS6) was 31% with a median duration of response of 9 months (4–18+months).
Median OS (mOS) was 15.9 months, supporting potential benefit of this approach despite unremarkable outcomes
based on radiographic endpoints. It is possible that the use of PARP inhibition may have greater value when used in
conjunction with other therapies such as DNA-damaging agents. Potential benefit may also be more pronounced
if used earlier in the treatment course as has been the case with other treatments for high-grade gliomas.
In a study evaluating the newly emerging technology of BBB disruption, a phase I/IIa study evaluated an
implantable ultrasound device in patients with recurrent glioblastoma treated with carboplatin [16].Duringthe
dose escalation phase (n = 9), the chemotherapy dose was fixed, and the number of sonications (n = 3, 6 or 9) was
escalated with no dose-limiting toxicities detected. The expansion cohort of an additional 12 subjects treated with
nine sonications proved tolerable with one grade 3 wound-related toxicity. Postprocedural MRI demonstrated clear
4426 Future O ncol. (2021) 17(33) future science group
ASCO Neuro-Onc meeting report Conference Report
evidence of successful BBB disruption. Other systemic therapies and immunotherapies in conjunction with this
approach are currently being considered for clinical trials [17–19].
Advances in primary brain tumors: phase I trials
In the domain of adoptive immunotherapy, a phase I trial examined the safety profile of autologous yδT cells
administered intracranially via a Rickham catheter (placed intracavitarily) in addition to standard radiation and
chemotherapy in patients with newly diagnosed glioblastoma [20].yδT cells are rare immune cells that possess both
innate and adaptive immune properties. Among the six patients at dose level 1, all were IDH wildtype, and five
were MGMT unmethylated. One patient was unable to generate an adequate amount of yδT cells a potential
limitation of this type of approach. Toxicities were manageable with only one patient experiencing grade 3 adverse
events. Serum TNFαremained elevated through day 30 and beyond. This type of approach appears to be feasible
and well tolerated thus far. Additional correlative study results with dose escalation will be of interest.
Advances in primary brain tumors: case series
A multi-institutional case series of H3K27M diffuse gliomas were treated with the highly selective dopamine D2
receptor antagonist ONC201 was presented [21]. Partial responses (not centrally reviewed) in two of seven patients
raises interest for this type of approach especially in a patient population with limited treatment options. As proof
of concept has been demonstrated in only a small number of patients [22], studies such as this help lend further
support. Prospective trials (Clinicaltrials.gov: NCT03295396, NCT03416530 and NCT02525692) using this
approach are ongoing.
Advances in primary brain tumors: trials in progress
We highlight two ongoing clinical trials in which preliminary results are eagerly awaited. PVSRIPO (recombi-
nant oncolytic poliovirus) and Pembrolizumab in Patients with Recurrent Glioblastoma (LUMINOS-101) is an
ongoing phase II trial for recurrent glioblastoma [23]. The previous phase I trial of PVSRIPO as monotherapy
demonstrated safety and tolerability [24]. A number of key inclusion and exclusion criteria (enhancing tumor size
between 1 and 5.5 cm; exclusion of high-dose steroids, notable midline-crossing tumor or extensive subependymal
involvement) will need to be considered when interpreting the generalizability of the study results when available.
ThesecondisaphaseI/II study of the exportin-1 inhibitor selinexor for newly diagnosed or recurrent glioblastoma
in combination with current standard of care [21]. Blocking the transport of specific proteins out of the tumor cell
nucleus into the cytoplasm has long been an attractive target for the treatment of cancer [25]. In the ongoing study,
newly diagnosed glioblastoma patients all receive selinexor, and the MGMT-methylated glioblastoma patients also
receive radiation and temozolomide. The MGMT unmethylated subjects are treated with radiation but not the
temozlomide.
Advances in CNS metastatic tumors: CSF biomarkers
Our discussion of advances for CNS metastases is focused on the exciting work in the realm of CSF biomarkers. A
retrospective single-institution study was conducted evaluating CSF circulating tumor cells (CTCs) as a predictive
biomarker of benefit from proton craniospinal irradiation (CSI) for leptomeningeal metastases [26]. The patient
composition consisted of both lung (47%) and breast (38%) brain metastasis. Median OS was 8 months with
mPFS of 6 months, notably better than accepted historical comparisons, potentially due to selection bias. Low
baseline CSF CTCs were associated with a longer mOS, suggesting that this can be used as a biomarker to select
which patients should be offered more aggressive treatment strategies. It remains unclear, for now, as to how CSF
CTCs may guide on-treatment management of leptomeningeal metastases.
Another study evaluating CSF biomarkers examined circulating tumor DNA (ctDNA) in patients with triple-
negative breast cancer (TNBC) to evaluate their risk for the development of brain metastases [27].PatientsCSF
was analyzed for ctDNA after treatment with neoadjuvant chemotherapy. Thirty-nine percent of patients with
TNBC who underwent neoadjuvant chemotherapy were positive for ctDNA. Among these, 98.4% developed
radiographically evident brain metastases. Of the patients who were negative for CSF-ctDNA after treatment
(61%), the overwhelming majority (99%) did not develop brain metastases during the course of the study. Findings
from this study indicate that for patients with TNBC, positive CSF-ctDNA levels are associated with a higher
risk of recurrent brain metastases, poorer OS and poorer recurrence-free survival. Accordingly, this could influence
future science group www.futuremedicine.com 4427
Conference Report Baskaran, Kumthekar, Heimberger & Lukas
screening protocols and open up the possibility of prophylactic approaches to prevent brain metastases for this
patient population in the future.
Conclusion
Several new developments are underway for the treatment of primary CNS tumors and for the use of CSF
biomarkers in CNS metastases. Results from phase III trials for glioblastoma, including GEINO 1401 and EORTC
1709/CCTG CE.8, do not alter the standard of care but do help inform our understanding of the management of
this disease and future directions for investigation in the newly diagnosed setting. A nonrandomized phase II trial of
BRAF +MEK inhibitors in papillary BRAF V600E craniopharyngioma, on the other hand, demonstrated excellent
response rates in treatment-naive patients, influencing our therapeutic paradigm in these rare tumors. Studies of
CSF CTCs and CSF ctDNA help lay the groundwork for CSF studies and potentially important prognostic and
predictive biomarkers for CNS metastatic disease.
Acknowledgments
This meeting summary was presented at the Robert H. Lurie Comprehensive Cancer Center 2021 Oncology Review on 16 July
2021. The authors thank Lyndsey Van for assistance with manuscript preparation.
Financial & competing interests disclosure
The authors have no relevant afliations or nancial involvement with any organization or entity with a nancial interest in or
nancial conict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, hon-
oraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. NIH funding: Rimas V
Lukas, Priya Kumthekar, and Amy B Heimberger are all supported by: P50 CA221747 (CA/NCI NIH HHS/United States).
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as: of interest; •• of considerable interest
1. Domenech M, Fabregat-Franco C, Mesia C et al. Long-term results of the GEINO 1401 TRIAL: randomizing patients to stop or to
continue temozolomide until 12 cycles. J. Clin. Oncol. 39(Suppl. 15), 2013 (2021).
This negative phase III trial demonstrated lack of benefit from additional cycles of temozolomide for patients with newly
diagnosed glioblastoma, demonstrating stability of disease after six adjuvant cycles.
2. Blumenthal DT, Gorlia T, Gilbert MR et al. Is more better? The impact of extended adjuvant temozlomide in newly diagnosed
glioblastoma: a secondary analysis of EORTC and NRG Oncology/RTOG. Neuro On col. 19(8), 1119–1126 (2017).
3. Stupp R, Mason WP, van den Bent MJ et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J.
Med. 352(10), 987–996 (2005).
4. Hegi ME, Diserens AC, Gorlia T et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N.Engl.J.Med.
352(10), 997–1003 (2005).
5. Roth P, Gorlia T, Reijneveld JC et al. EORTC 1709/CCTG CE.8: a phase III trial of marizomib in combination with
temozolomide-based radiochemotherapy versus temozolomide-based radiochemotherapy alone in patients with newly diagnosed
glioblastoma. J. Clin. Oncol. 39(Suppl. 15), 2004 (2021).
This negative phase III trial demonstrated lack of benefit from adding the brain-penetrant proteasome inhibitor marizomib to
the standard of care treatment for newly diagnosed glioblastoma.
6. Roth P, Mason WP, Richardson PG, Weller M. Proteasome inhibition for the treatment of glioblastoma. Expert Opin. Invest. Drugs.
29(10), 1133–1141 (2020).
7. Brastianos PK, Twohy E, Geyer SM et al. Alliance A071601: phase II trial of BRAF/MEK inhibition in newly diagnosed papillary
craniopharyngiomas. J. Clin. Oncol. 39(Suppl. 15), 2000 (2021).
•• This is the first prospective clinical trial demonstrating substantial radiographic response in the majority of craniopharyngioma
patients receiving systemic therapy.
8. Brastianos PK, Shankar GM, Gill CM et al. Dramatic response of BRAF V600E mutant papillary craniopharyngioma to targeted
therapy. J. Natl Cancer Inst. 108(2), djv310 (2015).
9. Juratli TA, Jones PS, Wang N et al. Targeted treatment of papillary craniopharyngiomas harboring BAF V600E mutations. Cancer
125(17), 2910–2914 (2019).
10. Brastianos PK, Taylor-Weiner A, Manley PE et al. Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas. Nat.
Genet. 46(2), 161–165 (2014).
4428 Future O ncol. (2021) 17(33) future science group
ASCO Neuro-Onc meeting report Conference Report
11. Ahluwalia MS, Rauf Y, Li H et al. Randomized phase 2 study of nivolumab (nivo) plus either standard or reduced dose bevacizumab
(bev) in recurrent glioblastoma (rGBM). J. Clin. Oncol. 39(Suppl. 15), 2015 (2021).
12. Nayak L, Molinaro AM, Peters K et al. Randomized phase II and biomarker study pembrolizumab plus bevacizumab versus
pembrolizumab alone for patients with recurrent glioblastoma. Clin. Cancer Res. 27(4), 1048–1057 (2021).
13. Ducray F, Sanson M, Chinot OL et al. KS02.4. A olaparib in recurrent IDH-mutant high-grade glioma (OLAGLI). J. Clin. Oncol.
35(Suppl. 15), 2007 (2021).
14. Lu Y, Kwintkiewicz J, Liu Y et al. Chemosensitivity of IDH1-mutated gliomas due to an impairment in PARP-1 mediated DNA repair.
Cancer Res. 77(7), 1709–1718 (2017).
15. IDH-mutant tumors vulnerable to PARP inhibition. Cancer Discov. 7(4), OF4 (2017).
16. Idbaih A, Ducray F, Stupp R et al. A phase I/IIa study to evaluate the safety and efficacy of blood–brain barrier (BBB) opening with the
SonoCloud-9 implantable ultrasound device in recurrent glioblastoma patients receiving IV carboplatin. J. Clin. Oncol. 39(Suppl. 15),
2049 (2021).
This phase I/IIa trial of implantable ultrasound for blood–brain barrier disruption in recurrent glioblastoma demonstrated
feasibility, safety and evidence of blood–brain barrier disruption.
17. Zhang DY, Dmello C, Chen L et al. Ultrasound-mediated delivery of paclitaxel for glioma: a comparative study of distribution, toxicity,
and efficacy of albumin-bound versus cremaphor formulations. Clin. Cancer Res. 26(2), 477–486 (2020).
18. Sonabend AM, Stupp R. Overcoming the blood–brain barrier with an implantable ultrasound device. Clin. Cancer Res. 25(13),
3750–3752 (2019).
19. Sabbagh A, Beccaria K, Ling X et al. Opening of the blood–brain barrier using low-intensity pulsed ultrasound enhances responses to
immunotherapy in preclinical glioma models. Clin. Cancer Res. 27(15), 4325–4337 (2021).
20. Nabors LB, Lamb LS, Beelen MJ et al. Phase 1 trial of drug resistant immunotherapy: a first-in-class combination of MGMT-modified
γδ t cells and temozolomide chemotherapy in newly diagnosed glioblastoma. J. Clin. Oncol. 39(Suppl. 15), 2057 (2021).
21. Odia Y, Sumrall AL, Cloughesy TF et al. Single agent activity of ONC201 in non-midline H3 K27M-mutant diffuse gliomas. J. Clin.
Oncol. 39(Suppl. 15), e14037 (2021).
22. Chi AS, Tarapore RS, Hall MD et al. Pediatric and adult H3 K27M-mutant diffuse midline glioma treated with the selective DRD2
antagonist ONC201. J. Neurooncol. 145(1), 97–105 (2019).
23. Sloan AE, Buerki RA, Murphy C et al. LUMINOS-101: Phase 2 study of PVSRIPO with pembrolizumab in recurrent glioblastoma. J.
Clin. Oncol. 39(Suppl. 15), TPS2065 (2021).
24. Desjardins A, Gromeier M, Herndon JE 2nd et al. Recurrent glioblastoma treated with recombinant poliovirus. N. Engl. J. Med. 379(2),
150–161 (2018).
25. Gravina GL, Senapedis W, McCauley D et al. Nucleo-cytoplasmic transport as a therapeutic target of cancer. J. Hematol. Oncol. 7, 85
(2014).
26. Wijetunga NA, Boire AA, Yamada Y et al. Cerebrospinal fluid circulating tumor cells as a predictive biomarker for proton craniospinal
irradiation for leptomeningeal metastases. JClinOncol.39(Suppl. 15), 2011 (2021).
This study demonstrated the an association between cerebrospinal fluid circulating tumor cells and survival outcomes in patients
with solid tumor leptomeningeal metastases treated with craniospinal irradiation.
27. Raimondi L, Naso G, Lazzeroni R et al. Usefulness of assessment of circulating tumor DNA (ctDNA) of cerebrospinal fluid (CSF)
samples for early detection of brain metastasis (BrM) in patients with triple-negative breast cancer (TNBC). J. Clin. Oncol. 39(Suppl.
15), 507 (2021).
future science group www.futuremedicine.com 4429
ResearchGate has not been able to resolve any citations for this publication.
Article
507 Expression of Concern The abstract by Raimondi et al entitled, “Usefulness of assessment of circulating tumor DNA(ctDNA) of cerebrospinal fluid(CSF) samples for early detection of brain metastasis (BrM) in patients with triple-negative breast cancer (TNBC),” published in Journal of Clinical Oncology 39, no. 15_suppl (May 20, 2021) 507-507, is under further review. Questions have been raised regarding the integrity of the methods, results, and analysis of the reported abstract. Until the authors and their institutions can fully provide additional information, readers should interpret the findings presented with caution. An update will be provided when our investigation is complete. Background: Despite improvements in treatments, patients diagnosed with TNBC still have poor prognosis for a higher tendency of developing BrM. Identifying patients at high risk of BrM, enabling to predict who will take advantage from appropriate additional treatment, remains a critical problem. ctDNA represents a valuable tool associated with the outcome and the aggressiveness of breast cancer but no prognostic and predictive biomarker has been identified to predict the development of BrM in TNBC. We studied the usefulness of assessment of CSF-ctDNA for early identification of the risk of BrM in TNBC. Methods: Between January 2016 and December 2020, 323 newly diagnosed non-metastatic TNBC patients who underwent neoadjuvant therapy+surgery(NACT) with complete response(CR)were prospectively enrolled. After surgery, samples of CSF measuring ctDNA were obtained from all patients: CSF-ctDNA was extracted with the QIAamp Circulating Nucleic Acid Kit (Qiagen, Valencia, CA, USA) and ctDNA levels were measured. Survival curves were estimated using the Kaplan-Meier method and compared with the Log-rank test. Multivariate Cox regression was used to identify the risk of mortality at three years. Results: After NACT, CSF-ctDNA was detectable in 126/323 (39%) patients, 101/126 (80%) were diagnosed at III stage. 124 of 126 (98.4%) ctDNA+ patients subsequently developed BrM. In contrast, only 2 (2/197, 1%) ctDNA- patients subsequently developed BrM and the 195 other patients remain in a CR (p < 0.001, Fisher's exact test). CSF-ctDNA did associate with PFS and OS: undetectable ctDNA was associated with superior PFS (HR 0.3; p = 0.002) and OS (HR 0.2; p < 0.01), indicating survival is largely determined by the onset of BrM. With a median follow-up of 3 years, median PFS of ctDNA+ vs ctDNA- patients was 13 months vs not reach, p = 0.004 (by Log-rank test). Median OS for ctDNA+ vs ctDNA- patients was 16 months after NACT vs not reach, p = 0.0016 (by Log-rank test). At multivariate analysis detectable CSF-ctDNA emerged as the best predictor of the develop of BrM and 24-month mortality (HR:3.62; p < 0.0001). Age, stage, Ki67% and response to chemotherapy were not significantly associated with the prognosis. Conclusions: After NACT, detectable CSF-ctDNA significantly associates with PFS and OS, identifying early at-risk patients to develop BrM in TNBC.
Article
e14037 Background: H3 K27M-mutant diffuse midline glioma is an invariably lethal form of brain cancer that disproportionately affects children and young adults and has no effective treatment following front-line radiation. The initial disease definition in the 2016 WHO Classification of Tumors of the Central Nervous System regarded the H3 K27M mutation as pathognomonic, though the definition was updated in 2018 restricting the diagnosis to histologically diffuse gliomas that involve midline CNS structures (cIMPACT-NOW update 2). ONC201 is an investigational anti-cancer small molecule, DRD2 antagonist and ClpP agonist that has induced durable tumor regressions by RANO-HGG criteria in a registration cohort of recurrent diffuse midline glioma, H3 K27M-mutant patients treated with single agent ONC201. Methods: We present 7 patients with H3 K27M-mutant diffuse gliomas were enrolled in ONC201 clinical studies, though excluded from the registration cohort due to involvement of non-midline CNS structures, all within the cerebral hemispheres (3 frontal, 1 temporal, 1 frontotemporal, 1 parietal, and 1 corona radiata). Results: Two of the 7 patients underwent objective responses by RANO-HGG criteria as assessed by investigator, which was associated with clinical benefit that included increased mobility and level of alertness. Conclusions: These results demonstrate that H3 K27M-mutant diffuse gliomas occur outside of midline CNS structures, and suggest that ONC201 has single agent activity in H3 K27M-mutant gliomas irrespective of CNS location.
Article
2004 Background: Patients with newly diagnosed glioblastoma receive postoperative standard therapy with radiotherapy (RT), and concomitant and up to six cycles of maintenance temozolomide (TMZ) chemotherapy (TMZ/RT→TMZ). Marizomib is a novel, irreversible and brain-penetrant pan-proteasome inhibitor with encouraging findings in preclinical models and early-stage clinical trials for patients with newly diagnosed and recurrent glioblastoma. Therefore, a phase 3 trial was designed to explore the activity of marizomib in addition to TMZ/RT→TMZ. ClinicalTrials.gov Identifier: NCT03345095 Methods: EORTC 1709/CCTG CE.8 is a multicenter, randomized, controlled, open label phase 3 superiority trial. Eligibility criteria included histologically confirmed newly diagnosed glioblastoma and a Karnofsky performance status (KPS) > 70. Eligible patients were stratified for institution, age, KPS as well as extent of surgery, and centrally randomized in a 1:1 ratio. The primary objective of this study is to compare overall survival (OS) in patients receiving marizomib in addition to standard treatment with patients receiving standard treatment only. Secondary endpoints include progression-free survival (PFS), safety, neurocognitive function, and quality of life. Results: The study was opened at 49 EORTC sites in Europe, 23 CCTG sites in Canada, and 8 sites in the US. Patient enrolment started in June 2018 and was close to completion at the time of a planned interim analysis in September 2020. A total of 749 patients (of the planned 750) were randomized when the IDMC recommended to discontinue enrollment. Age, KPS and extent of resection were well balanced between the 2 study arms. No difference in median OS was observed between the standard arm (15.9 months) and the marizomib arm (15.7 months; HR = 0.99). Median PFS was 6.1 vs. 6.2 months (HR = 1.02). Patients in the marizomib group had more often grade 3/4 treatment-emergent adverse events (TEAE) compared to the standard therapy group (42.6% vs. 20.5%), including ataxia, hallucinations and headache. Conclusions: The addition of marizomib to standard radiochemotherapy did not improve OS or PFS in patients with newly diagnosed glioblastoma. Final survival analyses including determination of MGMT promoter methylation status and analyses of other secondary endpoints are ongoing. Clinical trial information: NCT03345095.
Article
2049 Background: Low intensity pulsed ultrasound (LIPU) in conjunction with intravenous microbubbles can transiently and reversibly disrupt the blood-brain barrier (BBB), allowing for an increase in the tissue concentration of chemotherapy agents in the brain. Mass spectrometry data from preclinical models (mouse, swine) showed a > 5x enhancement in carboplatin brain concentrations, which correlated well with the spatial distribution of a Gadolinium (Gd) contrast agent used for magnetic resonance imaging (MRI). Methods: The primary objective of this phase I/IIa study (NCT03744026) was to demonstrate the safety of BBB disruption using LIPU in patients with recurrent glioblastoma. This study was a 3+3 design using escalating numbers (3, 6, 9) of activated 1 MHz ultrasound emitters. Nine patients were treated in the escalation phase and another 12 patients were treated with 9 emitters in the expansion phase. Eligibility included recurrent GBM (any recurrence) with a maximum tumor size of < 70 mm. The SonoCloud-9 device (CarThera, Paris, France) was implanted during tumor debulking/resection surgery and replaced the bone flap, with the device targeting the tumor and surrounding peritumoral brain. The device was activated every four weeks for a duration of 270 seconds, concomitantly with IV DEFINITY microbubbles (10 ml/kg), to disrupt the BBB prior to administration of carboplatin (AUC 4-6). MRI was performed to verify safety and evaluate efficacy of BBB disruption with Gd enhancement. Results: No DLTs were observed. The overall tolerance of the SonoCloud-9 implant was good, with two transient, manageable grade 3 wound infections and one grade 1 acquired meningocele event considered as probably related to the overall procedure. The most frequent neurologic adverse events were grade 1 blurred vision (5%) and dizziness (5%). Conclusions: Significant Gd enhancement was observed after more than 90% of sonication sessions, suggesting effective BBB disruption and carboplatin enhancement. Clinical trial information: NCT03744026.
Article
2007 Background: There is a need to develop new treatments in IDH-mutant high-grade gliomas recurring after radiotherapy and chemotherapy. Based on preclinical studies showing that IDH-mutant tumors could be vulnerable to PARP inhibition we launched a phase II study to test the efficacy of olaparib (Lynparza) monotherapy in this population. Methods: Adults with recurrent high-grade IDH-mutant gliomas after radiotherapy and at least one line of alkylating chemotherapy (PCV or TMZ), KPS > 60, normal organ function were enrolled. The primary endpoint was 6 months PFS according to RANO criteria. Patients were treated with olaparib 300 mg twice daily. We used a single-stage Fleming design with p0 = 30%, p1 = 50%, a type I unilateral error rate of 5% and a power of 80%. Results: 35 patients with recurrent IDH-mutant gliomas (IDH1R132H-mutant n = 32, other IDH mutation n = 3, 1p/19 codeleted n = 16, 1p/19q non-codeleted n = 14) were enrolled (malignantly transformed low-grade gliomas n = 21, anaplastic gliomas n = 8, glioblastomas n = 6). Median time since diagnosis was 7.4 years (1-22 years), median time since radiotherapy was 2.8 years (0.6-18 years), median number of previous chemotherapy lines was 2 (1-5). With a median follow-up of 11 months, 30 patients had stopped treatment due to tumor progression and 2 patients were still on treatment 16 to 18 months after treatment start. At 6 months, 11/35 patients were progression-free (31 %). According to RANO criteria, based on local investigator analysis, 2 patients (5%) had a partial response and 14 patients a stable disease (37%) with a median duration of response of 9 months (4-18 months+). Median PFS and OS were 2.3 and 15.9 months and were similar in 1p/19q codeleted and non-codeleted patients. A grade 3 olaparib-related adverse event was observed in 5 patients (14%, lymphopenia n = 3, fatigue n = 2, diarrhea n = 1) and a grade 2 in 15 patients (43%), most frequently consisting in fatigue (23%), gastrointestinal disorders (20%) and lymphopenia (20%). No patient definitively stopped olaparib due to side effects. Conclusions: In this heavily pre-treated population of recurrent IDH-mutant gliomas, olaparib monotherapy was well tolerated and resulted in some activity supporting its evaluation in association with alkylating chemotherapy in recurrent IDH-mutant gliomas in future studies. Clinical trial information: NCT03561870.
Article
2013 Background: We previously presented our results of the GEINO 1401 trial that randomized patients diagnosed with glioblastoma and treated with chemoradiotherapy and adjuvant temozolomide (TMZ) followed by six cycles of TMZ, to receive an extended use of TMZ up to 12 cycles or to control. We found no differences in 6-months neither progression free survival (PFS) nor overall survival (OS). In this report we actualize our results and analyse long-term survivor patients (LTSP). Methods: The trial NCT02209948 randomized (ratio 1:1) 159 patients diagnosed with glioblastoma who had been treated with standard therapy to stop treatment or to continue up to 12 cycles of TMZ. Patients were stratified based on their O6-methylguanine-DNA-methyltransferasa (MGMT) methylation status and presence or absence of measurable disease at inclusion. We update here OS outcomes and analyse the data of LTSP defined as an OS over 30 months from diagnosis. Results: At a median follow-up of 20 months, 82.4% of the patients had died and 89.9% had progressed. The median OS from randomization was 22.0 months for the control arm and 18.2 for the experimental arm: HR 0.957 (95%CI 0.806-1.136, p = 0.615). At 2 years from randomization there were a 61% of survivors in the TMZ group and 62% in the control group. There were a 49.7% of LTSP showing no differences between TMZ and control group. We found a higher prevalence of methylated MGMT in LTSP, but no differences were shown in patients with or without measurable disease at inclusion, status of IDH and the use of bevacizumab after progression. Conclusions: Adding 6 cycles of TMZ after the first 6 adjuvant cycles confers no additional benefit in OS. Nearly 50% of the patients included in GEINO 1401 who had been previously treated with TMZ 6 cycles without progressing were LTSP. Clinical trial information: NCT02209948.
Article
2015 Background: Trials with anti-PD1 in rGBM have shown limited efficacy. VEGF is highly up regulated proangiogenic growth factor in GBM contributing to tumor-associated immunosuppression. Preclinical data suggests a potential dose effect of anti-VEGF therapy on immunomodulation. Hence, a combination of anti-PD1 and anti-VEGF may be a promising approach in rGBM. Methods: 90 patients with first-recurrent GBM were randomized (1:1) to nivolumab (240 mg IV Q2 weeks) with bevacizumab at standard (10 mg/kg; Arm A) or at low dose (3 mg/kg; Arm B) IV Q2 weeks. Stratification included extent of resection, age, performance status and MGMT methylation status. Single cell RNA sequencing with CITE-seq was used to analyze blood samples from pre- and 8 weeks post-treatment among 8 responders and 8 non-responders. Progression-free survival (PFS) and overall survival (OS) were compared between two arms. Results: 90 patients (Median age 60.6 years ranged 27.4-86.4, 67.8% male, median KPS 80) were enrolled between May 2018 and Jan 2020. Patients were followed in median 7.7 months (Range 0.7, 28.2). 35 of 88 patients were MGMT methylated (2 indeterminate). Overall OS was not significantly different between arm A and arm B (1 year: 41.1 vs 37.7%, p = 0.14), while OS was better for arm A in age > 60 (At 1-year: 46.2% vs 23.8%; Median: 10.6 vs 5.9 months; P = 0.046). OS was no different in the two arms for age ≤ 60 years (At 1-year: 35.6% vs 56.4; Median 8.0 vs 12.4 months; P = 0.90). Single cell RNA sequencing with CITE-seq was used to analyze blood samples from 16 patients, baseline and 8 weeks post treatment. Standard dose bevacizumab treated patients had decreased myeloid derived suppressor cells and an inflammatory response gene signature at 8 weeks. Most frequent toxicities ( > 20%) included fatigue (45.6%), proteinuria (34.4 %), diarrhea (28.9%), hypertension (23.3%) and lipase increase (21.1%). Toxicities in grade 3-4 were hypertension (7.8%), fatigue (5.6) and other non-neurological toxicities including DVT, PE, infection, and abnormal liver function. Conclusions: Overall PFS and OS rates appear similar for nivolumab with either standard or low-dose bevacizumab compared to historical benchmarks of bevacizumab monotherapy. Nivolumab with standard bevacizumab may benefit older but not younger patients. Ongoing response evaluation and immunocorrelative data will be presented. Clinical trial information: NCT03452579.
Article
2000 Background: Craniopharyngiomas, a rare brain tumor along the pituitary-hypothalamic axis, can cause significant clinical sequelae. Surgery and radiation, the only effective treatments, can cause significant morbidity. Genetic analysis of craniopharyngiomas revealed that 95% of papillary craniopharyngiomas (PCP) have BRAF V600E mutations (Brastianos et al. Nature Genetics 2014). We evaluated the efficacy of BRAF/MEK inhibition in patients (pts) with previously untreated PCP. Methods: Eligible pts without prior radiation whose PCP screened positively for BRAF mutations were treated with oral vemurafenib/cobimetinib in 28-day cycles. The primary endpoint of response rate (RR) based on centrally determined volumetric data was evaluated in 16 pts, where a partial response was defined as >20% decrease in volume. This single arm, Simon two-stage phase 2 trial had 89% power to detect a true RR of at least 30% (vs. the null RR 5%; alpha=0.04). In this design, 3 or more observed volumetric responses in 16 evaluable pts would be considered promising activity. Results: In the 16 pts evaluated, 56% were female, and the median age was 49.5 years. Median follow-up was 22 months (95% CI: 16-26.5) and median number of treatment cycles was 8. Three patients progressed after therapy was discontinued and none have died. Based on volumetric response criteria, 14 of 15 pts with volumetric data available for central review had response to therapy (93%; 95% CI: 68% to 99.8%). Of 16 patients evaluable based on local review, 15 had response to therapy (93.75%; 95% CI: 70% to 99.8%). The median tumor reduction was -83% (range: -52% to -99%). The one nonresponder received 2 days of treatment before coming off therapy due to toxicity. Median progression-free survival was not reached. Grade 3 toxicities at least possibly related to treatment occurred in 12 pts (rash in 6 pts). Grade 4 toxicities were observed in two pts: hyperglycemia (n=1) and increased CPK (n=1). Three pts discontinued treatment for adverse events. Conclusions: Vemurafenib/cobimetinib resulted in an objective response in all pts who received 1 or more cycles of therapy. Our study indicates that BRAF/MEK inhibitors could be a powerful tool in the treatment of previously untreated PCP and warrants further evaluation in larger studies. A second arm of this study is enrolling pts with progressive PCP after prior radiotherapy. Support: U10CA180821, U10CA180882; U24CA196171, U10CA180868 (NRG); Genentech; https://acknowledgments.alliancefound.org. Clinical trial information: NCT03224767.
Article
2011 Background: Leptomeningeal metastasis (LM) involves seeding of tumor cells to the cerebrospinal fluid (CSF) and the leptomeninges. Proton craniospinal irradiation (pCSI) has been shown to be potentially effective for patients with solid tumor LM. We evaluated whether CSF circulating tumor cells (CSF-CTC) and neuroimaging correlate with outcomes in patients with LM treated with pCSI. Methods: We reviewed a single-institution retrospective database of patients treated with pCSI for LM between 2018-2020 who had ≥ 3 months (mos.) follow-up and identified 58 patients. Pre-pCSI CSF-CTC using CellSearch and magnetic resonance imaging (MRI) data, and post-pCSI CSF-CTC nadir before initiation of new cancer-directed therapy were assessed. The optimal cutoff for pre-pCSI CSF-CTC was determined using maximally selected rank statistics. Kaplan Meier analysis was used to identify univariate correlates with CNS progression free survival (CNS PFS) and overall survival (OS), calculated from start of pCSI. Multivariate Cox proportional hazards modeling was used to test independence of univariate associations. Results: The median follow-up for patients who were censored (n = 15, 26%) was 15 mos. (interquartile range (IQR): 9 -21). Most patients were diagnosed with lung (n = 27, 47%) or breast cancer (n = 22, 38%). The median CNS PFS and OS were 6 mos. (IQR: 3 – 9) and 8 mos. (IQR: 5 – 18), respectively. Of the 49 patients with pre-pCSI CSF-CTCs analyzed, CSF-CTCs were identified in 43 (88%). Pre-pCSI CSF-CTC < 53/3mL was associated with improved CNS PFS (11.8 vs 6.0 mos., p = 0.01), and a trend toward improved OS (16.7 vs 7.7 mos., p = 0.08). On pre-pCSI MRI, patients with parenchymal brain metastases (n = 33, 57%) had worse OS (6.7 vs 12.7 mos., p = 0.01) but not CNS PFS. Patients with both brain and spine LM (n = 42, 72%) compared to those only one site or no visible disease (n = 16, 28%) showed worse CNS PFS (5.8 vs 7.5 mos., p = 0.03) and OS (7.7 vs 16.7 mos., p = 0.05). In a multivariate model, pre-pCSI CSF-CTC was significantly associated with CNS PFS (p = 0.03) while brain and spine LM on MRI was not (p = 0.20) No patient had an increase in CSF-CTC immediately post-pCSI, and in those with both detectable pre-pCSI CSF-CTCs and a post-pCSI measurement (n = 29, 50%), the median decrease at nadir was 37/3mL (range: 0-200) occurring at a median of 1.6 mos. (range: 0.5 -5.2). A decrease in CSF-CTC > 37/3mL was associated with improved CNS PFS (7.1 vs 4.4 mos., p = 0.04) but not OS (12.5 vs.7.7 mos., p = 0.2). Conclusions: Proton CSI is an effective treatment for patients with solid tumor LM and can result in prolonged disease control in some patients. Lower CSF-CTC count prior to pCSI and larger changes after pCSI are predictive of survival outcomes, arguing for early pCSI intervention for solid tumor LMD. Early treatment escalation after pCSI can be considered for patients with high pre-pCSI CSF-CTC and a smaller nadir post-pCSI.
Article
2057 Background: Temozolomide (TMZ) transiently upregulates NKG2D ligands targeted by innate immune effector cells. Lymphodepletion impairs this immune response, however, genetic modification of ex vivo expanded γδ T cells with an MGMT-expressing lentivector confers resistance to TMZ, allowing concurrent chemotherapy and γδ T cell infusion, thereby targeting the tumor when NKG2DL are maximally expressed. This Drug Resistant Immunotherapy (DRI) is currently being evaluated in a Phase 1 first in human study (NCT04165941) and interim safety and biologic correlative analysis are detailed here for the first dosing cohort. Methods: Adults with newly diagnosed, untreated glioblastoma (GBM), adequate organ function, and a KPS≥70% will be enrolled. Subjects undergo subtotal resection and placement of a Rickham reservoir followed 3-4 weeks by apheresis from which γδ T cells are expanded, transduced with an MGMT-expressing lentivector, harvested, and cryopreserved. Standard of care induction TMZ/radiation therapy is initiated followed by 6 cycles of maintenance TMZ. Intravenous TMZ (150mg/m ² ) and intracranial dosing of 1 x 10 ⁷ γδ T cells occur on day 1 of each maintenance cycle. Daily oral TMZ 150mg/m ² follows for Days 2-5. Dose level 1 (DL1) subjects receive 1 fixed dose of γδ T cells and DL2 receive 3 doses administered on Day 1 of each of first 3 cycles of TMZ dependent on absence of dose limiting toxicity. Primary endpoint is safety; secondary endpoints include progression free and overall survival. Immunologic and genomic correlative analyses are being conducted at specific time points from peripheral blood and cerebral spinal fluid collected from the Rickham. Results: Six subjects (4 females, 2 males) have been enrolled in DL1. All subjects were IDH1-WT with 5 subjects MGMT unmethylated and 1 methylated. Of these, 1 generated inadequate gd T cells and 2 withdrew consent prior to DRI treatment. For the 3 that received DRI, treatment-related adverse events with maximum CTCAE Grade 3 occurred in 1 subject; UTI, dehydration, and thrombocytopenia. The most common Grade 1/2 events included: fever, leukopenia, nausea, and vomiting which were attributable to TMZ or radiotherapy. Circulating T cells remained below normal range throughout maintenance phase in 2/3 subjects. NK and gd T cell numbers remained within low normal range for 3/3 and 2/3 subjects, respectively. Serum Th1 (IFNg, IL-2, TNFa) and Th2 (IL4, IL5, IL-10) cytokines were within clinical range although TNFa remained elevated from the gdT cell infusion through day +30 in 2 subjects. Conclusions: Administration of MGMT-gene modified gdT cells and TMZ as DRI is feasible in lymphodepleted subjects during TMZ maintenance phase and sufficiently safe to warrant further investigation at additional doses. Clinical trial information: NCT04165941.