Comprehensive genomic profiling (CGP) and the subsequent discussions in molecular tumor boards (MTBs) are becoming an integral part of personalized cancer care. The patient with metastatic renal cell carcinoma (mRCC) presented here demonstrated an absence of a favorable response accompanied by adverse events after receiving dual immunotherapy with nivolumab plus ipilimumab in combination with a poly (adenosine diphosphate–ribose) polymerase inhibitor, niraparib. This determination was made based on the initial CGP report and the initial MTB. Following the progression of the disease and the emergence of immune-related adverse events, a second CGP was conducted, and several subsequent MTBs were held. The decision was made to transition the patient’s treatment to temsirolimus plus bevacizumab, with the rechallenge of immunotherapy with pembrolizumab. The response evaluation revealed a complete radiographic and molecular response. This case study underscores the mounting significance of precision oncology in the management of mRCC, thereby suggesting that mammalian target of rapamycin inhibitor may augment the efficacy of immunotherapy in select patients based on their genomic findings. A digital poster of this case is included in the supplemental materials.

Renal cell carcinoma (RCC) is a type of kidney cancer that generally manifests as a tumor in the proximal convoluted tubule of one kidney.[1] Treatment for this specific form of cancer can vary depending on several factors and may include surgery, radiation therapy, targeted therapy, and immunotherapy.[2] Comprehensive genomic profiling (CGP) has made a significant contribution to the field of cancer care by providing insights into cancer biology, facilitating the detection of multiple biomarkers, identifying actionable alterations, and ultimately improving patient outcomes.[3] This case report describes a patient with metastatic renal cell carcinoma (mRCC) who initially did not respond to dual immunotherapy (nivolumab and ipilimumab) combined with the poly (adenosine diphosphate–ribose) polymerase inhibitor (niraparib) and experienced persistent immune-related adverse events (ir-AEs). Following a series of molecular tumor boards (MTBs), the patient exhibited a positive response to a novel combination of pembrolizumab, temsirolimus, and bevacizumab without notable AEs, achieving a complete radiographic and molecular response with minimal residual disease (MRD)–negative status that remains ongoing at 12 months. To the best of our knowledge, this case study is the first to demonstrate how CGP and MTBs were used to personalize cancer treatment for a patient with mRCC, leading to a successful rechallenge with immunotherapy in combination with angiogenesis and mammalian target of rapamycin (mTOR) inhibitors. The patient consented to publication of this case.

A 35-year-old asymptomatic woman with RCC and lung and bone metastases was admitted without prior treatment. The CGP using FoundationOne CDx revealed a tumor mutational burden of 1 mutation per megabase, microsatellite stability, BAP1 Y241fs1, TSC2 splice site 774 + 2T>A, and TP53 R249fs94 alterations, with a PD-L1 combined positive score of 2% and tumor proportion score of 2% by immunohistochemistry (Table 1). Following a comprehensive evaluation of the patient’s genetic profile by oncologists and cancer scientists in the MTB, and after discussing treatment options with the patient and obtaining consent for off-label treatment, she received dual immunotherapy (nivolumab 400 mg once monthly, ipilimumab 50 mg every 4–8 weeks) along with a PARPi (niraparib 100 mg/d). The treatment was initiated in March 2023, and by October of the same year, stable disease had been achieved. However, niraparib was discontinued in October 2023 due to dyspeptic symptoms. Following dual immunotherapy, an endoscopy revealed grade 1 gastritis, which led to the discontinuation of immunotherapy. However, the patient’s symptoms subsequently worsened. Fifteen days later, an endoscopy was performed, which revealed the presence of grade 3 gastritis, accompanied by symptoms such as nausea, vomiting, and weight loss. Consequently, the therapeutic regimen was discontinued. The patient was initiated on a treatment plan consisting of prednisone (1 mg/kg) and supportive care for a period of 5 months. During this time, new liver lesions were treated with radiofrequency ablation. A subsequent CGP test (Tempus|xT; Tempus Labs) was performed on a lymph node biopsy, revealing the same alterations in the BAP1 and TP53 genes as previously identified, but this time no TSC2 alteration was found. Following national and virtual international MTBs, the decision was made to proceed with rechallenge immunotherapy, switching the agent from nivolumab to pembrolizumab (200 mg every 3 weeks). This switch occurred 1 month after the discontinuation of steroids. The patient had discontinued steroids during the last month of the 5-month supportive care period and had received steroids for approximately 3 months. In light of the potential for undetected TSC2 alterations due to the heterogeneous nature of the tumor, temsirolimus (an initial dose of 10 mg and a maintenance dose of 15 mg administered intravenously per week) was recommended. Furthermore, bevacizumab (400 mg administered every 3 weeks) was added to the treatment regimen, given that TP53 alterations might have played a role in elevated vascular endothelial growth factor (VEGF) levels. In March 2024, the treatment was reinitiated. After a period of 2 months, positron emission tomography (PET) and computed tomography (CT) scans revealed the disappearance of the bone and mediastinal lesions, indicating a near-complete response (May 2024). However, the first MRD (Signatera; Natera) result was positive (May 2024). Consequently, the patient underwent continuous treatment, which led to a sustained complete response, as evidenced by the most recent combined PET-CT scan conducted in July 2024 (Fig. 1) and the second MRD test result becoming negative, indicating that the patient’s condition remains favorable with no evidence of residual disease (July 2024). Furthermore, a third MRD test performed in September 2024 also yielded a negative result, thereby confirming the maintenance of a complete molecular response (Fig. 2). This outcome indicates a noteworthy response to the treatment, with no evidence of residual disease in the most recent evaluations (MRD, December 2024, and combined PET-CT scan, January 2025) (see Figs. 1 and 2). The patient’s clinical history, treatments, and evaluation timeline are meticulously delineated (Fig. 3).

Figure 1

Patient scans showing liver parenchymal lesions (arrows) before (A) and after (B) treatment course. Thorax CT scans before treatment (C and F) and after 3 months (D and G) and 7 months (E and H) of treatment. PET scans before treatment (I) and after 3 months (J), 7 months (K), and 10 months (L) of treatment.

Figure 1

Patient scans showing liver parenchymal lesions (arrows) before (A) and after (B) treatment course. Thorax CT scans before treatment (C and F) and after 3 months (D and G) and 7 months (E and H) of treatment. PET scans before treatment (I) and after 3 months (J), 7 months (K), and 10 months (L) of treatment.

Close modal
Figure 2

Patient’s MRD follow-up.

MRD: minimal residual disease; MTM mean tumor molecules.

Figure 2

Patient’s MRD follow-up.

MRD: minimal residual disease; MTM mean tumor molecules.

Close modal
Figure 3

Patient’s treatment timeline.

CGP: comprehensive genomic profiling; IO: immuno-oncology therapy; ir-AE: immune-related adverse event; MTB, molecular tumor board; MTM: mean tumor molecules; PARPi: poly (adenosine diphosphate–ribose) polymerase inhibitor; RCC: renal cell carcinoma.

Figure 3

Patient’s treatment timeline.

CGP: comprehensive genomic profiling; IO: immuno-oncology therapy; ir-AE: immune-related adverse event; MTB, molecular tumor board; MTM: mean tumor molecules; PARPi: poly (adenosine diphosphate–ribose) polymerase inhibitor; RCC: renal cell carcinoma.

Close modal

We present the first case of complete radiologic and molecular response in a patient with mRCC using precision oncology principles, with rechallenge immunotherapy in combination with temsirolimus and bevacizumab after several MTBs. Combination therapies, including antiangiogenic agents, tyrosine kinase inhibitors, and mTOR inhibitors as well as immune checkpoint inhibitors (ICIs), have become the gold standard for mRCC treatment due to their synergistic effects on tumor progression.[4] Despite the encouraging outcomes observed in clinical trials, the response to treatment varies among patient subgroups. Furthermore, the real-world RCC population is heterogeneous, and it is possible that some subgroups will not benefit from these therapies.[5] In the treatment of intermediate- or poor-risk mRCC, dual immunotherapy is regarded as the standard of care.[6] This approach is hypothesized to yield a superior response due to the presence of BAP1 and other alterations that generate an immunogenic microenvironment.[7,8] On the other hand, BAP1 mutations have been identified as potential predictive biomarkers for the efficacy of immunotherapy. Patients with BAP1 mutations have demonstrated a remarkable response to immunotherapy, with significant clinical benefits being observed. Research has indicated that BAP1-associated signatures may offer superior predictive value for immunotherapy outcomes in comparison with other treatments, such as VEGFR and mTOR inhibitors.[7] On the other hand, the use of a PARPi has been rationalized by the BAP1 gene alteration[9] and the objective of enhancing the efficacy of immunotherapy.[10] Studies also show that PARPi and ICI combinations are effective with manageable toxicity in non-RCC cancers.[11] Two considerations provide the rationale for rechallenging this patient with immunotherapy. First, the patient’s disease was stabilized during the initial immunotherapy treatment. Second, a BAP1 alteration was detected in the patient’s genomic profile. This alteration is associated with a favorable predictive outcome in patients with RCC treated with immunotherapy.[7] Therefore, we hypothesize that immunotherapy may still be a suitable option. However, we also predicted that there was an alteration in the patient’s genomic profile that may reduce the response to immunotherapy. Consequently, we postulate that immunotherapy in combination with other agents may be more beneficial. The process of rechallenging ICIs in the aftermath of severe ir-AEs is a multifaceted and intricate procedure that necessitates a comprehensive, multidisciplinary approach.[12] Despite the risk of ir-AEs, some patients may derive significant clinical benefit from immunotherapy. The decision to rechallenge is usually based on a careful assessment of risks versus potential benefits.[13] In addition, recent advancements in the management of ir-AEs have enhanced the capacity to control these adverse events, thereby rendering rechallenge a feasible option for certain patients.[13] A study by Eldani et al[14] demonstrated that 73% of patients exhibited no recurrence following a rechallenge with ICIs, 13% experienced a recurrence, and 13% developed new ir-AEs. These findings suggest that it may be a safe approach, particularly after ir-AE regression and corticosteroid withdrawal.[14] A case study showed that a metastatic melanoma patient with immune reactions to nivolumab and ipilimumab tolerated pembrolizumab with hydrocortisone pretreatment.[15] Another study reported life-threatening ir-AEs in 0.2%, 0.4%, 2.9%, and 5.9% of patients with renal and colorectal cancers treated with pembrolizumab, nivolumab, ipilimumab, and the combination of ipilimumab and nivolumab, respectively.[16] Furthermore, a preclinical study demonstrated that dual PD-1 and CTLA-4 blockade led to a 77% inhibition of tumor growth in TSC2 loss cases, which was associated with an increase in tumor-infiltrating CD8+ and CD4+ T cells and a decrease in T regulatory cells, granulocyte-like myeloid derived suppressor cells, and regulatory CD11b+ DCs.[17] In light of these findings, a decision was made to proceed with an immunotherapy rechallenge, operating under the supposition that immunotherapy might still prove to be of benefit.

The PI3K/AKT/mTOR pathway, which plays a role in numerous biological processes, is found to be hyperactivated by TSC1 and TSC2 loss-of-function mutations. These mutations, which have been identified in over 40 tumor types, have the potential to serve as a prognostic indicator, suggesting an individual patient’s response to mTOR inhibitors.[18] A phase II study demonstrated a low response rate of 7% to everolimus in a pan-cancer cohort of patients with mTOR pathway alterations. Notably, two patients achieved objective responses; one of them had uterine cancer with biallelic TSC2 inactivating mutations.[19] Additionally, a phase II study indicated that alterations in AKT1, TSC1, and TSC2 genes and CTNNB1 mutations may serve as predictors of clinical benefit from temsirolimus treatment. Furthermore, CTNNB1 alterations have been associated with a prolonged progression-free survival.[20] In contrast, the RECORD-3 trial demonstrated that mutations in TSC1, TSC2, and mTOR did not have a significant effect on progression-free survival in patients with RCC treated with everolimus.[21] A study that received FDA approval demonstrated that temsirolimus enhanced median overall survival and progression-free survival by 49% among 626 patients diagnosed with advanced RCC. However, the study population was not selected based on specific gene alterations, and therefore it did not account for the potential effect on subgroups of patients with particular gene alterations.[22] The use of mTOR inhibitors is a double-edged sword, because they can have both immunosuppressive and immunostimulatory effects.[23] In a case of advanced, poorly differentiated thyroid cancer that progressed after lenvatinib and nivolumab plus ipilimumab with PTEN and TP53 mutations, treatment with temsirolimus in combination with nivolumab plus ipilimumab dual immunotherapy resulted in significant clinical improvement and disease control for approximately 6 months.[24]

Another case study demonstrated an almost complete response to PD-1 blockade in a mesenchymal tumor. The subsequent identification of a PTEN mutation indicated a potential resistance to immune checkpoint therapy, suggesting that the mTOR pathway might contribute to immune resistance.[25]

The identification of a TP53 alteration is another salient issue in this patient. A pan-cancer study demonstrated that mutation of the TP53 gene serves as a marker for VEGF overexpression, a pivotal protein involved in angiogenesis, the process by which new blood vessels form to supply the tumor with nutrients and oxygen.[26] Furthermore, studies have shown that the administration of VEGF and VEGFR inhibitors leads to a higher response rate among patients with a TP53 mutation.[27] In renal cell carcinoma, VEGF plays a critical role in tumor growth and metastasis by promoting the formation of new blood vessels that supply the tumor with nutrients and oxygen. Bevacizumab is a monoclonal antibody and effectively inhibits VEGF, depriving the tumor of these essential resources and thereby slowing its growth and spread.[28] In addition, the administration of combination therapy has been demonstrated to induce a transformation of a low immunoreactive tumor into a hot tumor.[29] The combination of bevacizumab and ICIs has been the subject of investigation as a potential treatment for various types of cancer, including RCC.[29] Moreover, a multitude of studies have demonstrated that the combination of bevacizumab and the mTOR inhibitor temsirolimus exhibits antitumor activity and is well tolerated.[30]

This case report is subject to certain limitations, including a relatively small sample size. On the other hand, the alteration in the TSC2 gene has not yet been characterized in any of the relevant databases. However, given that the c.774 + 1G>T alteration is reported to be pathogenic, it is likely that the c.774 + 2G>T alteration also results in dysfunction of the TSC2 tumor suppressor gene. Although temsirolimus and bevacizumab enhanced the efficacy of immunotherapy, their individual efficacy in this case remains unclear and requires further study.

This case study documented a patient with high-risk mRCC who exhibited an inadequate response to the combination of dual immunotherapy and PARPi initially recommended by the MTB. Subsequent MTBs provided critical insights that led to a reevaluation of the patient’s clinical status and genomic profiling. This process resulted in a successful rechallenge with immunotherapy combined with angiogenesis and mTOR inhibitors. Although these agents are approved as monotherapies for mRCC, this represents the first case report of a triplet combination guided by a specific genomic profile, which makes it a significant case study. However, the efficacy of this triple therapy is not yet fully understood, and further research is needed to elucidate the mechanisms by which mTOR inhibitors and immunotherapy work together. The paucity of literature on the personalized treatment of treatment-refractory or treatment-naive patients with mRCC who have refused standard therapy represents a knowledge gap that this case study aims to address. In addition, it may be useful for further refinement of subgroup analyses for personalized treatment approaches in patients with mRCC and specific genetic alterations.

Supplemental materials are available online with the article.

1.
Padala
SA,
Barsouk
A,
Thandra
KC,
et al
Epidemiology of renal cell carcinoma
.
World J Oncol
.
2020
;
11
:
79
87
.
2.
Escudier
B,
Porta
C,
Schmidinger
M,
et al
Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
.
2019
;
30
:
706
720
.
3.
Teuwen
LA,
Roets
E,
D’Hoore
P,
et al
Comprehensive genomic profiling and therapeutic implications for patients with advanced cancers: the experience of an academic hospital
.
Diagnostics
.
2023
;
13
:
1619
.
4.
Motzer
R,
Alekseev
B,
Rha
SY,
et al
Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma
.
N Engl J Med
.
2021
;
384
:
1289
1300
.
5.
Rossi
E,
Bersanelli
M,
Gelibter
AJ,
et al
combination therapy in renal cell carcinoma: the best choice for every patient
?
Curr Oncol Rep
.
2021
;
23
:
147
.
6.
Motzer
RJ,
Tannir
NM,
McDermott
DF,
et al
Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma
.
N Engl J Med
2018
;
378
:
1277
1290
.
7.
Liu
K,
Huang
Y,
Xu
Y,
et al
BAP1-related signature predicts benefits from immunotherapy over VEGFR/mTOR inhibitors in ccRCC: a retrospective analysis of JAVELIN Renal 101 and checkmate-009/010/025 trials
.
Cancer Immunol Immunother
.
2023
;
72
:
2557
2572
.
8.
Tokat
ÜM,
Adibi
A,
Aydın
E,
et al
Personalized immunotherapy achieves complete response in metastatic adenoid cystic carcinoma despite lack of conventional biomarkers
.
Curr Oncol
.
2024
;
31
:
5838
5849
.
9.
George
TJ,
Lee
JH,
DeRemer
DL,
et al
Phase II trial of the PARP inhibitor, niraparib, in BAP1 and other DNA damage response pathway-deficient neoplasms
.
JCO Precis Oncol
.
2024
;
8
:
e2400406
.
10.
Wanderley
CWS,
Correa
TS,
Scaranti
M,
et al
Targeting PARP1 to enhance anticancer checkpoint immunotherapy response: rationale and clinical implications
.
Front Immunol
.
2022
;
13
:
816642
.
11.
Drew
Y,
Kim
JW,
Penson
RT,
et al
Olaparib plus durvalumab, with or without bevacizumab, as treatment in PARP inhibitor-naïve platinum-sensitive relapsed ovarian cancer: a phase II multi-cohort study
.
Clin Cancer Res
.
2024
;
30
:
50
62
.
12.
Hu
H,
Wang
K,
Jia
R,
et al
Current status in rechallenge of immunotherapy
.
Int J Biol Sci
.
2023
;
19
:
2428
2442
.
13.
Dolladille
C,
Ederhy
S,
Sassier
M,
et al
Immune checkpoint inhibitor rechallenge after immune-related adverse events in patients with cancer
.
JAMA Oncol
.
2020
;
6
:
865
871
.
14.
Eldani
C,
Kostine
M,
Faure
M,
et al
Safety of immune checkpoint inhibitor rechallenge after severe immune-related adverse events: a retrospective analysis
.
Front Oncol
.
2024
;
14
:
1403658
.
15.
Cruse
AR,
Basilio
AJ,
Lawson
DH.
Metastatic melanoma treatment with pembrolizumab after hypersensitivity reaction to nivolumab: a case report
.
Case Rep Oncol
.
2023
;
16
:
1232
1236
.
16.
Davis
EJ,
Salem
JE,
Young
A,
et al
Hematologic complications of immune checkpoint inhibitors
.
Oncologist
.
2019
;
24
:
584
588
.
17.
Liu
HJ,
Lizotte
PH,
Du
H,
et al
TSC2-deficient tumors have evidence of T cell exhaustion and respond to anti-PD-1/anti-CTLA-4 immunotherapy
.
JCI Insight
.
2018
;
3
:
e98674
.
18.
Luo
C,
Ye
WR,
Shi
W,
et al
Perfect match: mTOR inhibitors and tuberous sclerosis complex
.
Orphanet J Rare Dis
.
2022
;
17
:
106
.
19.
Adib
E,
Klonowska
K,
Giannikou
K,
et al
Phase II clinical trial of everolimus in a pan-cancer cohort of patients with mTOR pathway alterations
.
Clin Cancer Res
.
2021
;
27
:
3845
3853
.
20.
Myers
AP,
Filiaci
VL,
Zhang
et al
Tumor mutational analysis of GOG248, a phase II study of temsirolimus or temsirolimus and alternating megestrol acetate and tamoxifen for advanced endometrial cancer (EC): an NRG Oncology/Gynecologic Oncology Group study
.
Gynecol Oncol
.
2016
;
141
:
43
48
.
21.
Voss
MH,
Chen
D,
Reising
A,
et al
PTEN expression, not mutation status in TSC1, TSC2, or mTOR, correlates with the outcome on everolimus in patients with renal cell carcinoma treated on the randomized RECORD-3 trial
.
Clin Cancer Res
.
2019
;
25
:
506
514
.
22.
Hudes
G,
Carducci
M,
Tomczak
P,
et al
Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma
.
N Engl J Med
.
2007
;
356
:
2271
2281
.
23.
El Hage
A,
Dormond
O.
Combining mTOR inhibitors and T cell‐based immunotherapies in cancer treatment
.
Cancers
.
2021
;
13
:
1359
.
24.
Oh
Y,
Park
JH,
Djunadi
TA,
et al
Deep response to a combination of mTOR inhibitor temsirolimus and dual immunotherapy of nivolumab/ipilimumab in poorly differentiated thyroid carcinoma with PTEN mutation: a case report and literature review
.
Front Endocrinol (Lausanne)
.
2024
;
15
:
1304188
.
25.
George
S,
Miao
D,
Demetri
GD,
et al
Loss of PTEN is associated with resistance to anti-pd-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma
.
Immunity
.
2017
;
46
:
197
204
.
26.
Li
AM,
Boichard
A,
Kurzrock
R.
Mutated TP53 is a marker of increased VEGF expression: analysis of 7,525 pan-cancer tissues
.
Cancer Biol Ther
.
2020
;
21
:
95
100
.
27.
Wheler
JJ,
Janku
F,
Naing
A,
et al
TP53 alterations correlate with response to VEGF/VEGFR inhibitors: implications for targeted therapeutics
.
Mol Cancer Ther
.
2016
;
15
:
2475
2485
.
28.
Qiao
T,
Zhao
J,
Xin
X,
et al
Combined pembrolizumab and bevacizumab therapy effectively inhibits non-small-cell lung cancer growth and prevents postoperative recurrence and metastasis in humanized mouse model
.
Cancer Immunol Immunother
.
2023
;
72
:
1169
1181
.
29.
Sepe
P,
Mennitto
A,
Corti
F,
Procopio
G.
Immunotherapeutic targets and therapy for renal cell carcinoma
.
Immunotargets Ther
.
2020
;
9
:
273
288
.
30.
Piha-Paul
SA,
Wheler
JJ,
Fu
S,
et al
Advanced gynecologic malignancies treated with a combination of the VEGF inhibitor bevacizumab and the mTOR inhibitor temsirolimus
.
Oncotarget
.
2014
;
5
:
1846
1855
.

Competing Interests

Source of Support: None. Conflicts of Interest: Razelle Kurzrock has received research funding from Boehringer Ingelheim, Debiopharm, Foundation Medicine, Genentech, Grifols, Guardant, Incyte, Konica Minolta, Medimmune, Merck Serono, Omniseq, Pfizer, Sequenom, Takeda, and TopAlliance and from the NCI, as well as consultant and/or speaker fees and/or advisory board/consultant for Actuate Therapeutics, AstraZeneca, Bicara Therapeutics, Inc., Biological Dynamics, Caris, Datar Cancer Genetics, Daïichi, EISAI, EOM Pharmaceuticals, lylon, LabCorp. Lanuaria, Merck, NeoGenomics, Neomed, Pfizer, Precirix, Prosperdx, Regeneron, Roche, TD2/Volastra, Turning Point Therapeutics, X-Biotech; has an equity interest in CureMatch Inc; serves on the Board of CureMatch and CureMetrix; and is a cofounder of CureMatch. The remaining authors have no disclosures.

Author notes

Cite as: Adibi A, Tokat ÜM, Aydın E, et al. Exceptional response in a patient with mRCC through precision-guided treatment involving immunotherapy rechallenge with temsirolimus and bevacizumab.

This work is published under a CC-BY-NC-ND 4.0 International License.

Supplementary data