Next Article in Journal
Advances in Genomic Discovery and Implications for Personalized Prevention and Medicine: Estonia as Example
Previous Article in Journal
Influence of the Depth of the Convolutional Neural Networks on an Artificial Intelligence Model for Diagnosis of Orthognathic Surgery
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Transient Response of Olaparib on Pulmonary Artery Sarcoma Harboring Multiple Homologous Recombinant Repair Gene Alterations

1
Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
2
ACT Genomics Co., Ltd., Taipei 114, Taiwan
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2021, 11(5), 357; https://doi.org/10.3390/jpm11050357
Submission received: 8 April 2021 / Revised: 24 April 2021 / Accepted: 27 April 2021 / Published: 29 April 2021
(This article belongs to the Section Mechanisms of Diseases)

Abstract

:
Primary pulmonary artery sarcoma (PPAS) is a rare malignancy arising from mesenchymal pulmonary artery cells and mimics pulmonary embolism. Palliative chemotherapy such as anthracycline- or ifosfamide-based regimens and targeted therapy are the only options. However, the evidence of clinically beneficial systemic treatment is scarce. Here, we report a case of disseminated PPAS achieving clinical tumor response to olaparib based on comprehensive genetic profiling (CGP) showing genetic alterations involving DNA repair pathway. This provides supportive evidence that olaparib could be a promising therapeutic agent for patients with disseminated PPAS harboring actionable haploinsufficiency of DNA damage repair (DDR).

1. Introduction

Primary pulmonary artery sarcoma (PPAS) is a rare malignancy, with an incidence rate of 1–30 per 100,000 people [1], which is originated from the pulmonary artery’s mesenchymal cells and mimicking pulmonary embolism [1,2,3]. Due to the ambiguous and rare manifestations of clinical image findings, PPAS is frequently misdiagnosed as various pulmonary thromboembolic diseases (e.g., chronic thromboembolic, pulmonary thromboembolism, or chronic thromboembolic pulmonary hypertension) [4]. As such, PPAS has a poor prognosis, with a median overall survival of less than 3 months without surgical resection and 1–2 years in the case with surgical resection [5,6].
Although surgical resection has been demonstrated to prolong survival [2,7], the immediate postoperative mortality rate has been reported to be ~13–15%, and surgical resection nearly never achieves R0 resection [3]. Furthermore, surgery has no role in disseminated PPAS. Unfortunately, palliative therapy (e.g., anthracycline-/ifosfamide-based chemotherapy regimens or targeted therapy) remains limited and unproven [8]. A clinically useful systematic targeted approach is crucial [9].
Herein, we present our experience of treating a patient with PPAS harboring MDM2 amplification; CDKN2A homozygous deletion; and RAD50, PTCH1, PTEN, CHEK1, MRE11, BRCA2, RB1, and BLM hemizygous deletion, treated with next-generation sequencing (NGS)-guided olaparib (Lynparza®) and achieving a transient partial response.

2. Case Report

A 71-year-old woman had progressively exertional dyspnea associated with productive cough and chest tightness for half a year. A computed tomography (CT) scan of the chest revealed a left main pulmonary arterial embolism and multiple metastatic nodules over both lung fields (Figure 1 and Figure 2). A CT-guided biopsy was performed for lung masses and pathology reported spindle-cell sarcoma, which was strongly positive for SMA, MDM2, and negative for cytokeratin (AE1/AE3), CAM5.2, CD117 (c-KIT), DOG-1, EMA (E29), P40, S-100, STAT6, TLE1, ER (6F11), PR (1A6), HMB-45, and CDK4. The positivity of SMA indicates muscular differentiation. After reviewing the CT scan and performing positron emission tomography (PET)/CT scan (Figure 2), disseminated PPAS with metastases to both lungs, the left brain, and the T6 vertebra was diagnosed. Palliative whole-brain radiotherapy was subsequently completed and systemic chemotherapy was suggested. However, the patient refused chemotherapy because of the poor performance status of 3 and her old age. As such, NGS-based comprehensive genetic profiling (CGP) was suggested and a lung tumor’s (70% tumor purity) formalin-fixed, paraffin-embedded (FFPE) specimen was sequenced using a targeted panel of 400+ cancer-related genes (ACTOnco®+) for molecular-matched therapeutic options [10].
The FFPE lung tumor was subject to next-generation sequencing (NGS) using the ACTOnco® + test (ACT Genomics, Taipei, Taiwan) to detect mutations in the coding region of 440 cancer-related genes and fusion of 31 genes. NGS was conducted at a mean depth of 1298x and 91% uniformity. Twelve nonsynonymous mutations were identified, while none of them was considered clinically significant (Table 1). The germline or somatic origin of these mutations cannot be determined due to lack of a paired normal tissue. No fusion gene was detected.
Clinically relevant copy number variants are shown in Table 2. Amplification of MDM2, classified as an oncogene, was identified and confirmed by immunohistochemical (IHC) staining, suggesting that MDM2 is a potential driver in sarcoma. MDM2 inhibitors may be the optimal treatment; however, they are not available outside clinical trials. The homozygous deletion of CDKN2A and hemizygous deletion of multiple tumor suppressor genes (TSGs) with haploinsufficiency, including RAD50 (chr 5), PTCH1 (chr 9), PTEN (chr 10), CHEK1 (chr 11), MRE11 (chr 11), BRCA2 (chr 13), RB1 (chr 13), and BLM (chr 15), were determined by observed copy number < 2 copies and zygosity of SNPs located on the genes (Figure 3). Further analysis showed loss of heterozygosity (LOH) of chromosome 11q, ranging from 11q13.1 to 11q24.3, suggesting single copy loss of another two DNA damage repair (DDR) genes, ATM and H2AX, that localize in this region. Although these DDR genes are all hemizygously deleted, multiple genetic lesions, alterations, and their haploinsufficient nature may lead to homologous recombination deficiency (HRD), as shown by LOH 55.5% of the genomic region interrogated. This indicates that the patient could benefit from a poly(ADP-ribose) polymerase (PARP) inhibitor.

3. Treatment Course

Based on the genomic alterations identified, olaparib 300 mg BID was prescribed for one week and the dose was reduced to 150 mg BID in the second week due to adverse events of grade 3 anorexia and grade 2 diarrhea. After a two-week treatment, the ECOG performance status improved from 3 to 1 and chest X-ray (CXR) revealed that the lung tumors remained stable (Figure 1). After one month of olaparib treatment, the patient developed a fever and progressive dyspnea, and chest CT revealed infiltrations over both lung fields, but the tumors decreased in size (Figure 1 and Figure 4). The primary and metastatic lung tumors decreased in diameter after olaparib treatment (Figure 4). Partial response was achieved according to the response evaluation criteria in solid tumors (RECIST) criteria. The symptoms of pneumonia improved after the completion of antibiotics. Unfortunately, her symptoms and performance status deteriorated after two months of olaparib treatment, and subsequent CXR confirmed tumor progression. Therefore, olaparib was discontinued, and hospice care was suggested. The patient died three months after the commencement of olaparib treatment.
This report was approved by the Institutional Review Board of Chang Gung Medical Foundation (202100118B0). Written consent was obtained from the patient’s legal guardian to use the images included in this report.

4. Discussion

In this case report, a patient with disseminated PPAS was treated with CGP-guided olaparib and achieved a transient partial response. Studies on CGP in PPAS are rare and only reported in abstracts [11,12]. In 21 cases of PPAS, notable alterations that may not be considered actionable included TP53 (47%), CDKN2A (36%), CDKN2B (25%), and RB1 (13%). In contrast, 11% of PPAS harbored clinically relevant genomic alterations that affected PDGFRA, RICTOR, CDK4, and KIT [11,12]. A total of 10 (48%) of PPAS harboring additional clinically relevant genomic alterations in EGFR, TSC2, ALK, and BRAF was considered actionable [11,12]. The mean tumor mutation burden (TMB) in the PPAS was 8.3 mutations per Mb (mut/Mb), with 14% of PPAS having TMB > 10 mut/Mb and 10% of PPAS having TMB > 20 mut/Mb, implying that some patients may benefit from immunotherapy [13,14]. No microsatellite instability-high (MSI-H) was found in nine cases with available results. However, no clinical benefit of targeted therapy or immunotherapy was reported based on NGS-based CGP in the literature.
In this case, several identified clinically relevant copy number variants provided the rationale for using targeted therapy (Table 2, Figure 5). All of the chromosomal locations and genes for those involved with hemizygous deletions are in Supplementary Table S1. The genomic amplification of the MDM2 gene in this tumor, as confirmed by IHC, might be one of the primary drivers [15]. MDM2 amplification was observed in 18.6% of sarcoma cases based on the TCGA database (PanCancer Atlas). Notably, MDM2 amplification is most common in well-differentiated liposarcoma/atypical lipomatous tumors, dedifferentiated liposarcoma, intimal sarcoma, and low-grade osteosarcoma [15].
Moreover, a homozygous deletion of CDKN2A was also found, implying that the p14/MDM2/p53 axis played an essential role in tumorigenesis in this particular case. MDM2 has been reported to be associated with tumorigenesis in multiple solid tumors and sarcoma, as it inhibits the function of p53 (a tumor suppressor) via ubiquitination, leading to proteasomal degradation. Targeting MDM2 alone or in combination with other agents could be a therapeutic strategy in cancer treatment [16,17]. However, MDM2 inhibitors are still under investigation in early clinical trials and are clinically unavailable [17,18]. In addition, the homozygous deletion of CDKN2A and hemizygous of RB1 were observed, and CDK4/6 inhibitors (palbociclib, ribociclib, and abemaciclib) may be an off-label option [19]. As a result of the cytostatic effects of CDK4/6 inhibitor monotherapy and insufficient clinical evidence, CDK4/6 inhibitors were not suggested for this patient.
Alternatively, the observed hemizygous deletions of several DDR genes (RAD50, BRCA2, BLM, CHEK1, MRE11, PTEN, ATM, and H2AX) might highlight a possible therapeutic route. According to Sanmartin et al., haploinsufficiency of DDR genes localized in 11q may confer higher sensitivity to olaparib treatment in neuroblastomas [20]. The collective haploinsufficiency from the 11q-loss would lead to a compromised DDR pathway. As DNA damage can occur at any time or position of the genome, the DDR pathway is essential to safeguard the genome’s integrity [21]. This repair mechanism can be exploited by cancer cells to facilitate the maintenance of cell viability, and a deficiency of this repair mechanism results in cancer susceptibility [22]. This deficiency also indicates the use of PARP inhibitors.
Clinical trials have demonstrated the effective induction of synthetic lethality in tumors harboring BRCA1/2 pathogenic mutations and the loss or disruption of crucial HRR genes [23,24,25]. The copy number loss of BRCAness genes such as MRE11, CHEK1, RAD51, PTEN deficiency; knockdown of RAD50; and BLM have all conferred sensitivity to PARP inhibitors across different tumor types [26,27,28,29,30,31,32]. Furthermore, PARP inhibitors significantly improved survival outcomes in several studies, both as monotherapy (e.g., TOPARP-B and QUADRA) [33,34] and combination therapy (e.g., PAOLA-1/ENGOT-ov25) [35]. These preclinical and clinical studies support expanding different PARP inhibitor treatments to patients with DDR-positive tumor types beyond those with BRCA mutations. Olaparib, an FDA-approved PARP inhibitor, has accumulated evidence establishing a relatively profound and sustained antitumor response across different tumor types, such as ovarian cancer, breast cancer, and pancreatic cancer, associated with germline BRCA1/2 mutations [36]. No documented PPAS cases harboring haploinsufficiency of DDR genes treated with PARP inhibitors have been reported to our knowledge. This case report provides a possible novel strategy for patients with such rare malignancies.

5. Conclusions

Given the lack of targeted therapies for patients with PPAS, olaparib could be a promising therapeutic agent for patients with disseminated PPAS harboring actionable haploinsufficiency of DDR genes. Future studies using NGS-based CGP to guide the treatment of PPAS-harboring haploinsufficiency of DDR genes are eagerly awaited.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/jpm11050357/s1, Table S1: Chromosomal locations and genes involved associated with hemizygous deletions in Figure 4.

Author Contributions

Conceptualization, C.-E.W.; methodology, C.-E.W., C.T.N. and K.T.T.; formal analysis, C.T.N. and K.T.T.; writing—original draft, C.-E.W. and C.T.N.; writing—review & editing, C.-E.W.; supervision, C.-E.W. All authors have read and agreed to the published version of the manuscript.

Funding

Chang Gung Memorial Hospital (CMRPG3K0601, CMRPG3J0971~2, and CRRPG3K0021) and Ministry of Science and Technology (109-2314-B-182A-148 -MY3, 109-2314-B-182-080-MY3).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of Chang Gung Medical Foundation (202100118B0).

Informed Consent Statement

Informed consent was obtained from the patient’s legal guardian to use the images included in this report.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to privacy or ethical issues.

Acknowledgments

This work was supported by grants from Linkou Chang-Gung Memorial Hospital (CMRPG3K0601, CMRPG3J0971~2, and CRRPG3K0021 to CEW) and the Ministry of Science and Technology (109-2314-B-182A-148-MY3, 109-2314-B-182-080-MY3 to CEW). The genome-wide LOH interpretation was carried out by Kuan-Ying Chen.

Conflicts of Interest

C.T.N. and K.T.T. are employees of ACT genomics, Co., Ltd.

References

  1. Miura, S.; Meirmanov, S.; Nakashima, M.; Hayashi, T.; Abe, K.; Tamaru, N.; Miyahara, Y.; Sekine, I. Intimal sarcoma of the pulmonary artery: Report of an autopsy case. Pathol Res. Pract. 2005, 201, 469–474. [Google Scholar] [CrossRef] [PubMed]
  2. Bandyopadhyay, D.; Panchabhai, T.S.; Bajaj, N.S.; Patil, P.D.; Bunte, M.C. Primary pulmonary artery sarcoma: A close associate of pulmonary embolism-20-year observational analysis. J. Thorac. Dis. 2016, 8, 2592–2601. [Google Scholar] [CrossRef] [Green Version]
  3. Wong, H.H.; Gounaris, I.; McCormack, A.; Berman, M.; Davidson, D.; Horan, G.; Pepke-Zaba, J.; Jenkins, D.; Earl, H.M.; Hatcher, H.M. Presentation and management of pulmonary artery sarcoma. Clin. Sarcoma Res. 2015, 5, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Lu, P.; Yin, B.B. Misdiagnosis of primary intimal sarcoma of the pulmonary artery as chronic pulmonary embolism: A case report. World J. Clin. Cases 2020, 8, 986–994. [Google Scholar] [CrossRef]
  5. Secondino, S.; Grazioli, V.; Valentino, F.; Pin, M.; Pagani, A.; Sciortino, A.; Klersy, C.; Callegari, M.G.; Morbini, P.; Dore, R.; et al. Multimodal Approach of Pulmonary Artery Intimal Sarcoma: A Single-Institution Experience. Sarcoma 2017, 2017, 7941432. [Google Scholar] [CrossRef] [Green Version]
  6. Mussot, S.; Ghigna, M.R.; Mercier, O.; Fabre, D.; Fadel, E.; Le Cesne, A.; Simonneau, G.; Dartevelle, P. Retrospective institutional study of 31 patients treated for pulmonary artery sarcoma. Eur. J. Cardiothorac. Surg. 2013, 43, 787–793. [Google Scholar] [CrossRef] [Green Version]
  7. Kruger, I.; Borowski, A.; Horst, M.; de Vivie, E.R.; Theissen, P.; Gross-Fengels, W. Symptoms, diagnosis, and therapy of primary sarcomas of the pulmonary artery. Thorac. Cardiovasc. Surg. 1990, 38, 91–95. [Google Scholar] [CrossRef] [PubMed]
  8. Judson, I.; Verweij, J.; Gelderblom, H.; Hartmann, J.T.; Schoffski, P.; Blay, J.Y.; Kerst, J.M.; Sufliarsky, J.; Whelan, J.; Hohenberger, P.; et al. Doxorubicin alone versus intensified doxorubicin plus ifosfamide for first-line treatment of advanced or metastatic soft-tissue sarcoma: A randomised controlled phase 3 trial. Lancet Oncol. 2014, 15, 415–423. [Google Scholar] [CrossRef]
  9. Van der Graaf, W.T.; Blay, J.Y.; Chawla, S.P.; Kim, D.W.; Bui-Nguyen, B.; Casali, P.G.; Schoffski, P.; Aglietta, M.; Staddon, A.P.; Beppu, Y.; et al. Pazopanib for metastatic soft-tissue sarcoma (PALETTE): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2012, 379, 1879–1886. [Google Scholar] [CrossRef]
  10. Merino, D.M.; McShane, L.M.; Fabrizio, D.; Funari, V.; Chen, S.J.; White, J.R.; Wenz, P.; Baden, J.; Barrett, J.C.; Chaudhary, R.; et al. Establishing guidelines to harmonize tumor mutational burden (TMB): In silico assessment of variation in TMB quantification across diagnostic platforms: Phase I of the Friends of Cancer Research TMB Harmonization Project. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef] [Green Version]
  11. Beaucaire-Danel, S.; Girard, N.; Elvin, J.A.; Suh, J.; Vergilio, J.A.; Ramkissoon, S.H.; Severson, E.A.; Daniel, S.; Killian, J.K.; Ali, S.M.; et al. Primary pulmonary sarcomas (PSRC): A comprehensive genomic profiling (CGP) study. J. Clin. Oncol. 2018, 36. [Google Scholar] [CrossRef]
  12. Ross, J.; Suh, J.; Ali, S.; Schrock, A.; Elvin, J.; Vergilio, J.; Ramkissoon, S.; Miller, V.; Stephens, P.J.; Gay, L. Pulmonary Sarcomas: A Comprehensive Genomic Profiling Study. J. Thorac. Oncol. 2017, 12, S2103. [Google Scholar] [CrossRef]
  13. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H., Jr.; et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: Prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020, 21, 1353–1365. [Google Scholar] [CrossRef]
  14. Samstein, R.M.; Lee, C.H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 2019, 51, 202–206. [Google Scholar] [CrossRef] [PubMed]
  15. Sciot, R. MDM2 Amplified Sarcomas: A Literature Review. Diagnostics 2021, 11, 496. [Google Scholar] [CrossRef]
  16. Wu, C.E.; Esfandiari, A.; Ho, Y.H.; Wang, N.; Mahdi, A.K.; Aptullahoglu, E.; Lovat, P.; Lunec, J. Targeting negative regulation of p53 by MDM2 and WIP1 as a therapeutic strategy in cutaneous melanoma. Brit. J. Cancer 2018, 118, 495–508. [Google Scholar] [CrossRef] [Green Version]
  17. Wu, C.E.; Pan, Y.R.; Yeh, C.N.; Lunec, J. Targeting P53 as a Future Strategy to Overcome Gemcitabine Resistance in Biliary Tract Cancers. Biomolecules 2020, 10, 1474. [Google Scholar] [CrossRef]
  18. Kocik, J.; Machula, M.; Wisniewska, A.; Surmiak, E.; Holak, T.A.; Skalniak, L. Helping the Released Guardian: Drug Combinations for Supporting the Anticancer Activity of HDM2 (MDM2) Antagonists. Cancers 2019, 11, 1014. [Google Scholar] [CrossRef] [Green Version]
  19. Pernas, S.; Tolaney, S.M.; Winer, E.P.; Goel, S. CDK4/6 inhibition in breast cancer: Current practice and future directions. Ther Adv. Med. Oncol. 2018, 10, 1758835918786451. [Google Scholar] [CrossRef] [Green Version]
  20. Sanmartin, E.; Munoz, L.; Piqueras, M.; Sirerol, J.A.; Berlanga, P.; Canete, A.; Castel, V.; Font de Mora, J. Deletion of 11q in Neuroblastomas Drives Sensitivity to PARP Inhibition. Clin. Cancer Res. 2017, 23, 6875–6887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Curtin, N.J. DNA repair dysregulation from cancer driver to therapeutic target. Nat. Rev. Cancer 2012, 12, 801–817. [Google Scholar] [CrossRef] [PubMed]
  22. Kotsantis, P.; Petermann, E.; Boulton, S.J. Mechanisms of Oncogene-Induced Replication Stress: Jigsaw Falling into Place. Cancer Discov. 2018, 8, 537–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mirza, M.R.; Monk, B.J.; Herrstedt, J.; Oza, A.M.; Mahner, S.; Redondo, A.; Fabbro, M.; Ledermann, J.A.; Lorusso, D.; Vergote, I.; et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N. Engl. J. Med. 2016, 375, 2154–2164. [Google Scholar] [CrossRef] [PubMed]
  24. Pujade-Lauraine, E.; Ledermann, J.A.; Selle, F.; Gebski, V.; Penson, R.T.; Oza, A.M.; Korach, J.; Huzarski, T.; Poveda, A.; Pignata, S.; et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017, 18, 1274–1284. [Google Scholar] [CrossRef] [Green Version]
  25. Oza, A.M.; Matulonis, U.A.; Malander, S.; Hudgens, S.; Sehouli, J.; del Campo, J.M.; Berton-Rigaud, D.; Banerjee, S.; Scambia, G.; Berek, J.S.; et al. Quality of life in patients with recurrent ovarian cancer treated with niraparib versus placebo (ENGOT-OV16/NOVA): Results from a double-blind, phase 3, randomised controlled trial. Lancet Oncol. 2018, 19, 1117–1125. [Google Scholar] [CrossRef]
  26. Coleman, R.L.; Oza, A.M.; Lorusso, D.; Investigators, A. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 1948. [Google Scholar] [CrossRef] [Green Version]
  27. Jiang, X.; Li, X.Y.; Li, W.H.; Bai, H.M.; Zhang, Z.Y. PARP inhibitors in ovarian cancer: Sensitivity prediction and resistance mechanisms. J. Cell Mol. Med. 2019, 23, 2303–2313. [Google Scholar] [CrossRef] [Green Version]
  28. Brandt, S.; Samartzis, E.P.; Zimmermann, A.K.; Fink, D.; Moch, H.; Noske, A.; Dedes, K.J. Lack of MRE11-RAD50-NBS1 (MRN) complex detection occurs frequently in low-grade epithelial ovarian cancer. BMC Cancer 2017, 17, 44. [Google Scholar] [CrossRef] [Green Version]
  29. Mateo, J.; Carreira, S.; Sandhu, S.; Miranda, S.; Mossop, H.; Perez-Lopez, R.; Rodrigues, D.N.; Robinson, D.; Omlin, A.; Tunariu, N.; et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N. Engl. J. Med. 2015, 373, 1697–1708. [Google Scholar] [CrossRef]
  30. Schuler, F.; Weiss, J.G.; Lindner, S.E.; Lohmuller, M.; Herzog, S.; Spiegl, S.F.; Menke, P.; Geley, S.; Labi, V.; Villunger, A. Checkpoint kinase 1 is essential for normal B cell development and lymphomagenesis. Nat. Commun. 2017, 8, 1697. [Google Scholar] [CrossRef]
  31. Piscitello, D.; Varshney, D.; Lilla, S.; Vizioli, M.G.; Reid, C.; Gorbunova, V.; Seluanov, A.; Gillespie, D.A.; Adams, P.D. AKT overactivation can suppress DNA repair via p70S6 kinase-dependent downregulation of MRE11. Oncogene 2018, 37, 427–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Zhang, M.; Liu, G.Y.; Xue, F.X.; Edwards, R.; Sood, A.K.; Zhang, W.; Yang, D. Copy number deletion of RAD50 as predictive marker of BRCAness and PARP inhibitor response in BRCA wild type ovarian cancer. Gynecol. Oncol. 2016, 141, 57–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Mateo, J.; Porta, N.; McGovern, U.; Elliott, T.; Jones, R.; Syndikus, I.; Ralph, C.; Jain, S.; Varughese, M.; Parikh, O.; et al. TOPARP-B: A Phase II Randomised Trial of the Poly(ADP)-Ribose Polymerase (PARP) Inhibitor Olaparib for Metastatic Castration Resistant Prostate Cancers (mCRPC) with DNA Damage Repair (DDR) Alterations. Brit. J. Cancer 2019, 121, 25. [Google Scholar] [CrossRef]
  34. Moore, K.N.; Secord, A.A.; Geller, M.A. Niraparib monotherapy for late-line treatment of ovarian cancer (QUADRA): A multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2019, 20, E242. [Google Scholar] [CrossRef]
  35. Grimm, C.; Cropet, C.; Ray-Coquard, I. Maintenance olaparib plus bevacizumab (bev) after platinum-based chemotherapy plus bev in patients (pts) with newly diagnosed advanced high-grade ovarian cancer (HGOC): Efficacy by timing of surgery and residual tumor status in the Phase III PAOLA-1 trial. Gynecol Oncol. 2020, 159, 19. [Google Scholar] [CrossRef]
  36. Jiang, X.; Li, W.H.; Li, X.Y.; Bai, H.M.; Zhang, Z.Y. Current status and future prospects of PARP inhibitor clinical trials in ovarian cancer. Cancer Manag. Res. 2019, 11, 4371–4390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Summary of the clinical course. D0 indicates the first day of olaparib treatment.
Figure 1. Summary of the clinical course. D0 indicates the first day of olaparib treatment.
Jpm 11 00357 g001
Figure 2. CT images (A,D) and PET/CT scans (B,C). (A,B) Primary pulmonary artery sarcoma (PPAS) with lung metastases by CT scan (A) and the corresponding PET/CT scan (B). (C) A whole-body PET scan showed PPAS with multiple metastases to the lung, bone, and brain. (D) Brain metastases on CT scan.
Figure 2. CT images (A,D) and PET/CT scans (B,C). (A,B) Primary pulmonary artery sarcoma (PPAS) with lung metastases by CT scan (A) and the corresponding PET/CT scan (B). (C) A whole-body PET scan showed PPAS with multiple metastases to the lung, bone, and brain. (D) Brain metastases on CT scan.
Jpm 11 00357 g002
Figure 3. Summary list of genes with single copy loss detected within chromosome 11q.
Figure 3. Summary list of genes with single copy loss detected within chromosome 11q.
Jpm 11 00357 g003
Figure 4. CT scan of the chest revealed a size reduction of tumors before (A,C,E) and after olaparib treatment (B,D,F). The asterisk (*) indicates a primary tumor, while the arrow symbol (→) indicates metastatic tumors. The primary tumor (*) decreased in diameter from 4.8 cm (A) to 3.0 cm (B) before and after olaparib treatment, respectively. The metastatic tumors of the lung (→) decreased in diameter from 2.0 cm (A), 3.4 cm (C), and 3.2 cm (E) to 0.8 cm (B), 1.6 cm (D), and 2.3 cm (F) before and after olaparib treatment, respectively. Partial response was achieved according to response evaluation criteria in solid tumors (RECIST) criteria.
Figure 4. CT scan of the chest revealed a size reduction of tumors before (A,C,E) and after olaparib treatment (B,D,F). The asterisk (*) indicates a primary tumor, while the arrow symbol (→) indicates metastatic tumors. The primary tumor (*) decreased in diameter from 4.8 cm (A) to 3.0 cm (B) before and after olaparib treatment, respectively. The metastatic tumors of the lung (→) decreased in diameter from 2.0 cm (A), 3.4 cm (C), and 3.2 cm (E) to 0.8 cm (B), 1.6 cm (D), and 2.3 cm (F) before and after olaparib treatment, respectively. Partial response was achieved according to response evaluation criteria in solid tumors (RECIST) criteria.
Jpm 11 00357 g004
Figure 5. Copy number profile of the lung biopsy sample. The red “dots” indicate amplified regions while the blue “dots” indicate deleterious regions.
Figure 5. Copy number profile of the lung biopsy sample. The red “dots” indicate amplified regions while the blue “dots” indicate deleterious regions.
Jpm 11 00357 g005
Table 1. Clinically relevant single nucleotide and small indel variants from FFPE lung biopsy.
Table 1. Clinically relevant single nucleotide and small indel variants from FFPE lung biopsy.
GeneChrExonAccession NumbercDNA ChangeAmino Acid ChangeCoverageAllele FrequencyCOSMIC ID
ADAMTS931NM_182920c.107C > TP36L114712.7%-
ATRXX9NM_000489c.1331G > AR444Q162617.7%COSM3424965
BRCA11710NM_007294c.2347A > GI783V130150.1%-
ERBB21727NM_004448c.3763G > TV1255L60047.0%COSM7313442
FLT3137NM_004119c.866A > CN289T196647.2%-
KDR48NM_002253c.983C > AP328H36939.6%COSM3825987
MUC16193NM_024690c.18096C > GH6032Q143863.4%-
NOTCH463NM_004557c.316C > AL106I164549.2%-
NSD154NM_022455c.1070A > GN357S119847.5%-
POLD11910NM_001256849c.1232A > CQ411P72648.5%-
RECQL48-NM_004260c.1391-4G > TSplice region149535.6%-
USH2A166NM_206933c.14404T > CS4802P197954.5%-
Table 2. Clinically relevant copy number variants (CNVs) from archival formalin-fixed, paraffin-embedded lung biopsy.
Table 2. Clinically relevant copy number variants (CNVs) from archival formalin-fixed, paraffin-embedded lung biopsy.
Pathway InvolvedGeneCopy NumberAlteration
MDM2-P53 pathwayMDM239Amplification
ARF-MDM2-P53 pathway; Rb-E2F-1 pathway; P16/Rb pathwayCDKN2A0Homozygous Deletion
Homologous RecombinationRAD50, BRCA2, BLM, CHEK1, MRE111Hemizygous Deletion
Nonhomologous End-joiningRB11Hemizygous Deletion
PTEN-PI3K-AKT pathwayPTEN1Hemizygous Deletion
Type I noncanonical Hh pathwayPTCH11Hemizygous Deletion
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wu, C.-E.; Ng, C.T.; Tan, K.T. Transient Response of Olaparib on Pulmonary Artery Sarcoma Harboring Multiple Homologous Recombinant Repair Gene Alterations. J. Pers. Med. 2021, 11, 357. https://doi.org/10.3390/jpm11050357

AMA Style

Wu C-E, Ng CT, Tan KT. Transient Response of Olaparib on Pulmonary Artery Sarcoma Harboring Multiple Homologous Recombinant Repair Gene Alterations. Journal of Personalized Medicine. 2021; 11(5):357. https://doi.org/10.3390/jpm11050357

Chicago/Turabian Style

Wu, Chiao-En, Ca Tung Ng, and Kien Thiam Tan. 2021. "Transient Response of Olaparib on Pulmonary Artery Sarcoma Harboring Multiple Homologous Recombinant Repair Gene Alterations" Journal of Personalized Medicine 11, no. 5: 357. https://doi.org/10.3390/jpm11050357

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

Article Metrics

Back to TopTop