Next Article in Journal
Identification of Schizophrenia Susceptibility Loci in the Urban Taiwanese Population
Previous Article in Journal
Exploring the Influence of Dark Triad and Light Triad Traits on Sport Sciences Students
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Adverse Events in Osimertinib Treatment for EGFR-Mutated Non-Small-Cell Lung Cancer: Unveiling Rare Life-Threatening Myelosuppression

1
The Legacy Heritage Cancer Center and Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel
2
Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
3
Department of Dermatology and Venereology, Emek Medical Centre, Afula 18341, Israel
4
Nutrition Unit, Galilee Medical Center, Nahariya 22000, Israel
*
Author to whom correspondence should be addressed.
Medicina 2024, 60(8), 1270; https://doi.org/10.3390/medicina60081270
Submission received: 11 July 2024 / Revised: 1 August 2024 / Accepted: 5 August 2024 / Published: 6 August 2024
(This article belongs to the Special Issue Recent Developments in Anticancer Drug Delivery)

Abstract

:
Recent advancements in targeted therapies for non-small-cell lung cancer (NSCLC), specifically focusing on epidermal growth factor receptor (EGFR) mutations, have revolutionized treatment strategies. Osimertinib, an approved therapy for metastatic NSCLC with EGFR mutations, highlights remarkable efficacy but also harbors the potential for severe adverse events, whose rarity or lack of precedence may mask their criticality. This article delves into the exploration of adverse events linked to osimertinib, shedding light on a rare yet life-threatening occurrence: severe myelosuppression. A case study detailing a patient with EGFR-mutated NSCLC exhibiting a robust treatment response but experiencing severe myelosuppression following osimertinib initiation is presented. Immediate discontinuation of osimertinib alongside concurrent blood transfusions facilitated toxicity recovery, prompting a successful reduction in myelosuppression severity upon re-administration at a lowered dosage.

1. Introduction

Lung cancer is responsible for causing the highest number of cancer-related deaths worldwide, where, in 2017, this cancer type caused more deaths than breast, prostate, colorectal, and brain cancers combined [1,2]. More than 80% of lung cancer cases are classified as non-small-cell lung cancer (NSCLC), with this cancer subtype being one of the main causes of the high lung cancer death rate [3,4]. NSCLC presents with few, if any, symptoms in its initial phases, resulting in most patients seeking treatment only once the disease has advanced and metastasized. This contributes significantly to a dismal patient prognosis [5].
When lung cancer is found to be mutated in the epidermal growth factor receptor (EGFR), targeted drugs of the tyrosine kinase inhibitor (TKI) family have proven to be successful treatment options. Multiple clinical studies have shown the superiority of EGFR TKIs as a first-line treatment [6,7,8] over chemotherapy, with a response rate of 70% [9]. There are currently five TKI drugs categorized into three generations that are available to treat EGFR-mutated NSCLC. First- and second-generation TKIs include erlotinib and gefitinib, and afatinib and dacomitinib, respectively, where osimertinib is a third-generation TKI. Osimertinib is the only agent with FDA approval to be utilized as adjuvant treatment for EGFR-mutated NSCLC [10].
Osimertinib, an irreversible, orally administered EGFR-TKI, was initially approved by the FDA in 2015 and was intended for use only as an adjuvant therapy after tumor resection in adult NSCLC patients with EGFR exon 19 deletions or exon 21 L858 mutations. In 2018, indications for osimertinib were expanded for use as a first-line therapy in adults with metastatic NSCLC whose tumors bare the aforementioned mutations. Osimertinib is also indicated for the treatment of metastatic NSCLC patients who test positive for the EGFR T790M mutation and whose disease has progressed during or following EGFR TKI therapy. Osimertinib covalently binds to certain mutant forms of EGFR, i.e., exon 19 deletion, L858R, and T790M, to prevent the activation of the EGFR-mediated cell signaling cascade. In this way, the drug works as an antagonist with antitumoral capabilities to potentially inhibit neoplasm growth in EGFR-overexpressing tumor cells and induce cell death [11,12].
In the FLAURA study for patients treated with osimertinib, several adverse events were reported. The most common side-effects included diarrhea (60%), rash or acne (59%), and nail effects (39%). Dry skin and stomatitis were also notable, affecting 38% and 29% of patients, respectively. Other frequently observed side-effects included decreased appetite (24%), cough (22%), and nausea (20%). Constipation, pruritus, and renal symptoms were reported in 18% of patients each. Fatigue and anemia affected 16% of patients, while dyspnea and vomiting were reported in 15% of patients. Headache, back pain, and upper respiratory tract infections occurred in 13% of patients. Pyrexia, insomnia, and nasopharyngitis were noted in 11% of patients each. Less common but significant effects included prolonged QT interval, increased aspartate aminotransferase, musculoskeletal pain, and alopecia [13].
Herein, we describe the first documented case of severe myelosuppression in a patient with metastatic non-small-cell lung cancer (NSCLC) harboring EGFR mutations. This life-threatening case condition occurred as a result of osimertinib treatment.

2. Case Report

In January 2020, a 60-year-old female referred to the emergency room of Soroka Medical Center due to a rash on her hands and face, accompanied by itching and weakness. She was an otherwise healthy non-smoker on no medication and with no family history of cancer. She was hospitalized for further investigations. Laboratory investigations, including a complete blood count, biochemical profile, rheumatologic tests, oncologic markers, tuberculosis tests, urine tests, and stool tests, were within the normal range. A multidisciplinary conference, including a dermatologist, rheumatologist, and neurologist, concluded that the patient presented with a clinical picture matching that of dermatomyositis. Therefore, a skin biopsy was performed where histopathological results supported a diagnosis of dermatomyositis. Chest radiography (CXR) showed a rounded opacity in the left upper lobe (LUL). For further investigation, the patient underwent a total body computed tomography scan (CT) that visualized an LUL mass with a diameter of 17 mm, as well as enlarged neck and mediastinal lymph nodes (left). Magnetic resonance imaging (MRI) of the head showed no evidence of brain metastases. A left supraclavicular lymph node biopsy confirmed metastatic adenocarcinoma of the lung (poorly differentiated; TTF-1-positive; P40-negative). Positron emission tomography–computed tomography (PET-CT) showed hyper-metabolic uptake in the LUL mass (diameter of 17 mm) and the neck and mediastinal lymph nodes (11 mm), as well as in a splenic mass (diameter of 12 mm). A clinical diagnosis of metastatic non-small-cell lung cancer (NSCLC) T1N3M1 stage IV was made. Further molecular analysis revealed the disease to be EGFR-positive (p.Glu746_Ser752delinsVal.), PDL1 > 50%, ROS1-negative, ALK-negative, and BRAF-negative.
Treatment with osimertinib was initiated in February 2020 at a standard dose of 80 mg once daily. A complete blood count was performed and was within the normal range at this time. A month later, the patient’s dermatological symptoms were completely diapered, and we came to the conclusion that the clinical picture of dermatomyositis was paraneoplastic syndrome; two months later, the patient underwent a follow-up PET-CT, which revealed a good response to treatment with relatively less uptake in the LUL mass, and treatment with osimertinib was continued. In December 2020, MRI and PET-CT follow-ups showed stable disease; however, due to arthralgia, fatigue, and weakness, a complete blood count was performed that revealed moderate anemia (9 gr/dL (normal 12–16)), leukopenia (1.83 × 103/uL (normal 4.8–10.8)), neutropenia (1.72 × 103/uL (normal 2–8 × 103/uL)), and moderate thrombocytopenia (76 × 103/uL (normal 130–140)). Ten months later (October 2021), the patient was admitted to the emergency department upon being found comatose at home. Blood tests revealed severe myelosuppression (anemia of 4.3 gr/dL, thrombocytopenia of 5 × 103/uL, leukopenia of 0.92 × 103/uL, and neutropenia of 0.52 × 103/uL (normal 2–8 × 103/uL)); the patient was therefore admitted into the intensive care unit (I.C.U). A bone marrow biopsy was performed that showed no metastasis or malignancy, with no other pathological findings present. Osimertinib treatment was suspended, the patient was administered platelets and erythrocytes, and prednisone treatment was initiated. After one week, the patient’s condition had improved significantly, osimertinib treatment was resumed as before, and she was sent home. By December 2021, upon weekly follow-up blood tests, the patient’s condition had improved and her complete blood count returned to an acceptable range with hemoglobin (10.1 gr/dL), neutrophils (3.2 × 103/uL), and platelets (46 K × 103/uL). The patient had mild myelosuppression that did not require medical intervention; she was asked to continue follow-ups for blood tests. During this period, we communicated with “AstraZeneca” who initially indicated no connection to the treatment, and the patient continued rigorous follow-ups with blood tests. Two months later (February 2022), however, the patient was again admitted to the I.C.U in a comatose state with a complete blood count revealing the return of severe myelosuppression with hemoglobin (6.2 gr/dL), leukopenia (1.2 × 103/uL), neutrophils (0.94 × 103/uL), and platelets (10 K × 103/uL). Osimertinib treatment was immediately suspended. Blood and platelet transfusions were performed. A bone marrow biopsy was performed that revealed no evidence of metastatic carcinoma or plasma cell neoplasm or lymphoma.
The myelosuppression was suspected to be a drug-related adverse effect and thus, upon release from the hospital, the patient was carefully monitored by weekly blood tests, while osimertinib treatment was resumed as before (80 mg daily). The weekly blood tests began to show the progressive return of myelosuppression. The osimertinib dosage was thus reduced to 80 mg every alternative day. The rate of progression of the myelosuppression decreased; however, it continued to worsen and, as a result, the osimertinib dose was decreased to 40 mg every second day.
In April 2022, the patient’s blood count showed an improvement in hemoglobin (10 gr/dL), leukopenia (1.2 × 103/uL), neutrophil (3.5 × 103/uL), and platelet (36 K × 103/uL) counts. We continued to monitor our patient with weekly blood tests, which consistently showed stability. This approach effectively reduced myelosuppression to Grade 1/2, with no further events of severe myelosuppression observed. At present (July 2024), the patient remains on osimertinib treatment at 40 mg every second day with grade 1/2 myelosuppression that is treated with blood transfusions as needed.

3. Discussion

Lung cancer is the most commonly occurring cancer globally with NSCLC accounting for the vast majority of cases and EGFR being one of the main driver genes [1,14]. Multiple randomized phase III studies found both first- and second-generation EGFR-TKIs, gefitinib, erlotinib, and afatinib to significantly improve overall survival in advanced EGFR-mutated NSCLC as compared to standard, platinum-based combination chemotherapy [15,16]. This resulted in EGFR-TKI being recommended as the first-line treatment for patients with advanced EGFR-mutated NSCLC [17,18]. It was shown, however, that the majority of patients on this initial treatment ultimately developed therapeutic resistance [19].
Osimertinib is an oral, third-generation EGFR-TKI recommended for use as a first-line therapy in adults with metastatic, EGFR-mutated NSCLC [12]. This was due to the results of the FLAURA study—a double-blind, phase 3 trial comparing first-line osimertinib to gefitinib or erlotinib in patients with EGFR-mutation-positive NSCLC—that osimertinib achieved first-line status and became listed as a priority recommendation by the National Comprehensive Cancer Network guidelines in 2019 [20,21]. Unlike prior EGFR-TKI generations, osimertinib selectively and irreversibly binds to EGFR-mutants inhibiting downstream signaling pathways and ultimately preventing tumor cell proliferation while promoting apoptosis [22]. Osimertinib has been shown to exert significant therapeutic results, especially in NSCLC patients with the T790M EGFR-associated mutation. The ADURA study revealed a 71% overall response rate (ORR) and 10.1-month progression-free survival (PFS) in T790M-mutant NSCLC patients [23]. It is important to note that when compared to first-generation TKIs, osimertinib shows a lower rate of grade 3 adverse events, indicative of an improved safety profile [20,24].
A 2020 systematic review and meta-analysis assessing the safety and efficacy of osimertinib in treating NSCLC based on overall survival (OS), PFS, treatment response, and adverse events (AEs), that included 47 studies, found osimertinib to be associated with an improved OS and PFS in all patients, including when compared to other EGFR-TKIs or chemotherapy. The incidence of severe AEs ranged from 0 to 5%, with the most common severe AE being pneumonia that occurred in 3% of patients [25]. Another retrospective multicenter study assessing the efficacy and safety of osimertinib in advanced or recurrent NSCLC at various treatment lines revealed a 26.8% incidence of adverse events of grade ≥ 3, with the most common being anemia and cutaneous toxicity (both at 5%). Neutropenia and grade 3 pneumonitis were observed in just a single patient each (2%) [26].
Previous research has categorized the tumor immune microenvironment (TIME) into three distinct phenotypes: inflamed, immune-excluded, and immune-desert. These phenotypes are known to impact the efficacy of immune checkpoint inhibitors (ICIs) in cancer treatment. Studies focusing on EGFR-positive tumors suggest that these tumors typically exhibit an uninflamed phenotype, which partly explains the limited effectiveness of ICIs in EGFR-positive patients [27,28,29].
Moreover, investigations into the tumor immune microenvironment during EGFR tyrosine kinase inhibitor (TKI) treatment have indicated that levels of immunosuppressive cells such as M2 macrophages and myeloid-derived suppressor cells (MDSCs) tend to increase following the development of resistance. Conversely, the levels of immune-active cells, including dendritic cells (DCs) and CD8+ T cells, often decrease after resistance emerges to EGFR-TKIs [29,30,31].
Osimertinib therapy may act as a bridge to allogeneic hematopoietic stem cell transplantation for patients with refractory acute myeloid leukemia. Interestingly, EGFR covalent inhibitors, like osimertinib, also show activity against stem and progenitor cells in chronic myeloid leukemia (CML). The introduction of imatinib and subsequent second-generation tyrosine kinase inhibitors (TKIs) has significantly transformed CML from a fatal disease into a manageable condition requiring lifelong therapy. Despite this progress, leukemia stem cells in CML can persist independently of BCR-ABL, leading to resistance to imatinib and presenting a major clinical challenge. Osimertinib’s selective cytotoxicity against CD34+ cells suggests that combining it with imatinib might offer a promising strategy to address BCR-ABL-independent mechanisms of resistance in CML [32,33,34].
Recently, it was reported that AML to anecdotal osimertinib monotherapy suggested significant antileukemia activity, particularly as these patients had experienced multiple rounds of chemotherapy or had a poor performance status precluding treatment with conventional induction regimens [32,35]. However, these findings are not directly applicable to our patient. Our patient had a very good performance status and initiated osimertinib therapy as a first-line treatment and has not undergone chemotherapy. Additionally, multiple bone marrow biopsies have been performed to rule out any second primary or hematologic malignancies.
Our patient who we described before had a serious adverse event of myelosuppression during treatment with a third-generation epidermal growth factor receptor tyrosine kinase inhibitor (osimertinib) (Figure 1, Figure 2, Figure 3 and Figure 4). This is, in very rare cases, known to cause aplastic anemia, as described in the literature, and symptoms of drug-induced myelosuppression have been found to develop from days to months after initiation of treatment with the offending drug [36].
To the best of our knowledge, this is the first reported case of an EGFR-mutated NSCLC patient receiving first-line osimertinib suffering from life-threatening myelosuppression as a severe AE.

4. Conclusions

Herein, we report the rare occurrence of life-threatening, severe myelosuppression as a direct AE of first-line osimertinib treatment in an EGFR-mutated NSCLC patient showing good treatment response. Following temporary osimertinib discontinuation and concurrent blood transfusions to allow for toxicity recovery, a reduction in osimertinib dosage to 40 mg every second day was successful in reducing the myelosuppression to grade 1/2. Osimertinib may be safely administered to maintain stable disease in a patient with concurrent low-grade myelosuppression receiving blood transfusions as needed.

Author Contributions

Conceptualization, W.S.; methodology, W.S.; software, W.S.; validation, W.S., A.A.J., Y.D., O.A.S., S.S., L.T., K.S., A.M. and A.Y.; formal analysis, W.S.; investigation, W.S., A.A.J., Y.D., O.A.S., S.S., L.T., K.S., A.M. and A.Y.; resources, W.S.; data curation, W.S.; writing—original draft preparation, W.S., A.A.J., Y.D., O.A.S., S.S., K.S., A.M. and A.Y.; writing—review and editing, W.S., A.A.J., Y.D., O.A.S., S.S., L.T., K.S., A.M. and A.Y.; visualization, W.S.; supervision, W.S.; project administration, W.S.; funding acquisition, W.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of Soroka Medical Center on clinical and epidemiological characteristics of patients with thoracic tumors. This study was approved by the Institutional Review Board of Soroka Medical Center (approval no. 0316 on 8 December 2022).

Informed Consent Statement

Patient consent was waived due to the determination that the waiver or alteration would not negatively impact the rights and well-being of the subjects.

Data Availability Statement

The data either reside within this article itself or can be obtained from the authors upon making a reasonable request.

Acknowledgments

The authors extend their thanks to the dedicated medical and non-medical staff, including oncologists, radiologists, nurses, and secretaries, for their tireless efforts and contributions.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  2. Shalata, W.; Zolnoorian, J.; Migliozzi, G.; Abu Jama, A.; Dudnik, Y.; Cohen, A.Y.; Meirovitz, A.; Yakobson, A. Long-Lasting Therapeutic Response following Treatment with Pembrolizumab in Patients with Non-Small Cell Lung Cancer: A Real-World Experience. Int. J. Mol. Sci. 2023, 24, 5938. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  3. Chen, Z.; Fillmore, C.M.; Hammerman, P.S.; Kim, C.F.; Wong, K.-K. Non-small-cell lung cancers: A heterogeneous set of diseases. Nat. Rev. Cancer 2014, 14, 535–546. [Google Scholar] [CrossRef] [PubMed]
  4. Szeto, C.H.; Shalata, W.; Yakobson, A.; Agbarya, A. Neoadjuvant and Adjuvant Immunotherapy in Early-Stage Non-Small-Cell Lung Cancer, Past, Present, and Future. J. Clin. Med. 2021, 10, 5614. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  5. Fischer, B.M.; Mortensen, J.; Hansen, H.; Vilmann, P.; Larsen, S.S.; Loft, A.; Bertelsen, A.K.; Ravn, J.; Clementsen, P.F.; Høegholm, A.; et al. Multimodality approach to mediastinal staging in non-small cell lung cancer. Faults and benefits of PET-CT: A randomised trial. Thorax 2011, 66, 294–300. [Google Scholar] [CrossRef] [PubMed]
  6. Wu, Y.-L.; Zhou, C.; Hu, C.-P.; Feng, J.; Lu, S.; Huang, Y.; Li, W.; Hou, M.; Shi, J.H.; Lee, K.Y.; et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): An open-label, randomised phase 3 trial. Lancet Oncol. 2014, 15, 213–222. [Google Scholar] [CrossRef] [PubMed]
  7. Yang, J.C.-H.; Wu, Y.L.; Schuler, M.; Sebastian, M.; Popat, S.; Yamamoto, N.; Zhou, C.; Hu, C.-P.; O’Byrne, K.; Feng, J.; et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): Analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol. 2015, 16, 141–151. [Google Scholar] [CrossRef] [PubMed]
  8. Nan, X.; Xie, C.; Yu, X.; Liu, J. EGFR TKI as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer. Oncotarget 2017, 8, 75712–75726. [Google Scholar] [CrossRef]
  9. Jänne, P.A.; Wang, X.; Socinski, M.A.; Crawford, J.; Stinchcombe, T.E.; Gu, L.; Capelletti, M.; Edelman, M.J.; Villalona-Calero, M.A.; Kratzke, R.; et al. Randomized phase II Trial of erlotinib alone or with carboplatin and paclitaxel in patients who were never or light former smokers with advanced lung adenocarcinoma: CALGB 30406 trial. J. Clin. Oncol. 2012, 30, 2063–2069. [Google Scholar] [CrossRef]
  10. FDA. FDA Approves Osimertinib as Adjuvant Therapy for Non-Small Cell Lung Cancer with EGFR Mutations 2020. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-osimertinib-adjuvant-therapy-non-small-cell-lung-cancer-egfr-mutations (accessed on 9 March 2023).
  11. Song, C.; Yang, X. Osimertinib-Centered Therapy Against Uncommon Epidermal Growth Factor Receptor-Mutated Non-Small-Cell Lung Cancer—A Mini Review. Front. Oncol. 2022, 12, 834585. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. CDERF Highlights of Prescribing Information for TAGRISSO (Osimertinib) 2018. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/208065s011lbl.pdf (accessed on 18 April 2018).
  13. Ramalingam, S.S.; Vansteenkiste, J.; Planchard, D.; Cho, B.C.; Gray, J.E.; Ohe, Y.; Zhou, C.; Reungwetwattana, T.; Cheng, Y.; Chewaskulyong, B.; et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2020, 382, 41–50. [Google Scholar] [CrossRef] [PubMed]
  14. Molina, J.R.; Yang, P.; Cassivi, S.D.; Schild, S.E.; Adjei, A.A. (Eds.) Non-Small Cell Lung Cancer: Epidemiology, Risk Factors, Treatment, and Survivorship; Mayo Clinic Proceedings; Elsevier: Amsterdam, The Netherlands, 2008. [Google Scholar]
  15. Rosell, R.; Carcereny, E.; Gervais, R.; Vergnenegre, A.; Massuti, B.; Felip, E.; Palmero, R.; Garcia-Gomez, R.; Pallares, C.; Sanchez, J.M.; et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012, 13, 239–246. [Google Scholar] [CrossRef]
  16. Han, J.-Y.; Park, K.; Kim, S.-W.; Lee, D.H.; Kim, H.Y.; Kim, H.T.; Ahn, M.J.; Yun, T.; Ahn, J.S.; Suh, C.; et al. First-SIGNAL: First-line single-agent iressa versus gemcitabine and cisplatin trial in never-smokers with adenocarcinoma of the lung. J. Clin. Oncol. 2012, 30, 1122–1128. [Google Scholar] [CrossRef]
  17. Lee, C.K.; Brown, C.; Gralla, R.J.; Hirsh, V.; Thongprasert, S.; Tsai, C.-M.; Tan, E.H.; Ho, J.C.-M.; Chu, D.T.; Zaatar, A.; et al. Impact of EGFR inhibitor in non–small cell lung cancer on progression-free and overall survival: A meta-analysis. J. Natl. Cancer Inst. 2013, 105, 595–605. [Google Scholar] [CrossRef]
  18. Reck, M.; Popat, S.; Reinmuth, N.; De Ruysscher, D.; Kerr, K.M.; Peters, S. Metastatic non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2014, 25, iii27–iii39. [Google Scholar] [CrossRef]
  19. Cortot, A.B.; Jänne, P.A. Molecular mechanisms of resistance in epidermal growth factor receptor-mutant lung adenocarcinomas. Eur. Respir. Rev. 2014, 23, 356–366. [Google Scholar] [CrossRef]
  20. Soria, J.-C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in untreated EGFR-mutated advanced non–small-cell lung cancer. N. Engl. J. Med. 2018, 378, 113–125. [Google Scholar] [CrossRef] [PubMed]
  21. Shalata, W.; Jacob, B.M.; Agbarya, A. Adjuvant Treatment with Tyrosine Kinase Inhibitors in Epidermal Growth Factor Receptor Mutated Non-Small-Cell Lung Carcinoma Patients, Past, Present and Future. Cancers 2021, 13, 4119. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  22. Jänne, P.A.; Yang, J.C.-H.; Kim, D.-W.; Planchard, D.; Ohe, Y.; Ramalingam, S.S.; Ahn, M.-J.; Kim, S.-W.; Su, W.-C.; Horn, L.; et al. AZD9291 in EGFR inhibitor–resistant non–small-cell lung cancer. N. Engl. J. Med. 2015, 372, 1689–1699. [Google Scholar] [CrossRef]
  23. Mok, T.S.; Wu, Y.-L.; Ahn, M.-J.; Garassino, M.C.; Kim, H.R.; Ramalingam, S.S.; Shepherd, F.A.; He, Y.; Akamatsu, H.; Theelen, W.S.M.E.; et al. Osimertinib or platinum–pemetrexed in EGFR T790M–positive lung cancer. N. Engl. J. Med. 2017, 376, 629–640. [Google Scholar] [CrossRef]
  24. Mountzios, G.; Planchard, D.; Metro, G.; Tsiouda, D.; Prelaj, A.; Lampaki, S.; Shalata, W.; Riudavets, M.; Christopoulos, P.; Girard, N.; et al. Molecular Epidemiology and Treatment Patterns of Patients With EGFR Exon 20-Mutant NSCLC in the Precision Oncology Era: The European EXOTIC Registry. JTO Clin. Res. Rep. 2022, 4, 100433. [Google Scholar] [CrossRef] [PubMed]
  25. Liu, J.; Li, X.; Shao, Y.; Guo, X.; He, J. The efficacy and safety of osimertinib in treating nonsmall cell lung cancer: A PRISMA-compliant systematic review and meta-analysis. Medicine 2020, 99, e21826. [Google Scholar] [CrossRef] [PubMed]
  26. Kishikawa, T.; Kasai, T.; Okada, M.; Nakachi, I.; Soda, S.; Arai, R.; Takigami, A.; Sata, M. Osimertinib, a third-generation EGFR tyrosine kinase inhibitor: A retrospective multicenter study of its real-world efficacy and safety in advanced/recurrent non-small cell lung carcinoma. Thorac. Cancer 2020, 11, 935–942. [Google Scholar] [CrossRef] [PubMed]
  27. Dong, Z.Y.; Zhang, J.T.; Liu, S.Y.; Su, J.; Zhang, C.; Xie, Z.; Zhou, Q.; Tu, H.Y.; Xu, C.R.; Yan, L.X.; et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. OncoImmunology 2017, 6, e1356145. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, B.; Zhang, Y.; Zhao, J.; Wang, Z.; Wu, T.; Ou, W.; Wang, J.; Yang, B.; Zhao, Y.; Rao, Z.; et al. M2-polarized macrophages contribute to the decreased sensitivity of EGFR-TKIs treatment in patients with advanced lung adenocarcinoma. Med. Oncol. 2014, 31, 127. [Google Scholar] [CrossRef] [PubMed]
  29. Wang, C.; Fei, K.; Liu, L.; Duan, J.; Wang, Z.; Li, S.; Xu, J.; Zhang, X.; Tian, Y.; Qu, Y.; et al. Abnormal activation of NF-κB and MAPK signaling pathways affect osimertinib resistance and influence the recruitment of myeloid-derived suppressor cells to shape the immunosuppressive tumor immune microenvironment. Thorac. Cancer 2023, 14, 1843–1856. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  30. Feng, P.-H.; Yu, C.-T.; Chen, K.-Y.; Luo, C.-S.; Wu, S.M.; Liu, C.-Y.; Kuo, L.W.; Chan, Y.-F.; Chen, T.-T.; Chang, C.-C.; et al. S100A9+ MDSC and TAM-mediated EGFR-TKI resistance in lung adenocarcinoma: The role of RELB. Oncotarget 2018, 9, 7631–7643. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  31. Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef] [PubMed]
  32. Abraham, S.A.; Hopcroft, L.E.M.; Carrick, E.; Drotar, M.E.; Dunn, K.; Williamson, A.J.K.; Korfi, K.; Baquero, P.; Park, L.E.; Scott, M.T.; et al. Dual targeting of p53 and c-MYC selectively eliminates leukaemic stem cells. Nat. Cell Biol. 2016, 534, 341–346. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  33. Degterev, A.; Hitomi, J.; Germscheid, M.; Ch’En, I.L.; Korkina, O.; Teng, X.; Abbott, D.; Cuny, G.D.; Yuan, C.; Wagner, G.; et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol. 2008, 4, 313–321. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  34. Xia, L.; Liu, J.Y.; Yang, M.Y.; Zhang, X.H.; Jiang, Y.; Yin, Q.Q.; Luo, C.H.; Liu, H.C.; Kang, Z.J.; Zhang, C.T.; et al. Osimertinib Covalently Binds to CD34 and Eliminates Myeloid Leukemia Stem/Progenitor Cells. Cancer Res. 2024, 84, 479–492. [Google Scholar] [CrossRef] [PubMed]
  35. Kuntz, E.M.; Baquero, P.; Michie, A.M.; Dunn, K.; Tardito, S.; Holyoake, T.L.; Helgason, G.V.; Gottlieb, E. Targeting mitochondrial oxidative phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem cells. Nat. Med. 2017, 23, 1234–1240. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  36. Sala, L.; Mancin, M.; Pastore, A.; Seminati, D.; Cortinovis, D.; Bidoli, P.; Alberti, A. Aplastic anemia in a patient with advanced lung adenocarcinoma during first line osimertinib: A case report and literature review. Lung Cancer 2020, 142, 120–122. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Timeline of lung cancer EGFR mutation treatment (Tagrisso): phases, dose changes, and their effects on leukocyte (WBC) levels during follow-up (*, multiplication).
Figure 1. Timeline of lung cancer EGFR mutation treatment (Tagrisso): phases, dose changes, and their effects on leukocyte (WBC) levels during follow-up (*, multiplication).
Medicina 60 01270 g001
Figure 2. Timeline of lung cancer EGFR mutation treatment (Tagrisso): phases, dose changes, and their effects on platelet levels during follow-up (*, multiplication).
Figure 2. Timeline of lung cancer EGFR mutation treatment (Tagrisso): phases, dose changes, and their effects on platelet levels during follow-up (*, multiplication).
Medicina 60 01270 g002
Figure 3. Timeline of lung cancer EGFR mutation treatment (Tagrisso): phases, dose changes, and their effects on neutrophil levels during follow-up (*, multiplication).
Figure 3. Timeline of lung cancer EGFR mutation treatment (Tagrisso): phases, dose changes, and their effects on neutrophil levels during follow-up (*, multiplication).
Medicina 60 01270 g003
Figure 4. Timeline of lung cancer EGFR mutation treatment (Tagrisso): phases, dose changes, and their effects on hemoglobin levels during follow-up.
Figure 4. Timeline of lung cancer EGFR mutation treatment (Tagrisso): phases, dose changes, and their effects on hemoglobin levels during follow-up.
Medicina 60 01270 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shalata, W.; Abu Jama, A.; Dudnik, Y.; Abu Saleh, O.; Shalata, S.; Tourkey, L.; Sheva, K.; Meirovitz, A.; Yakobson, A. Adverse Events in Osimertinib Treatment for EGFR-Mutated Non-Small-Cell Lung Cancer: Unveiling Rare Life-Threatening Myelosuppression. Medicina 2024, 60, 1270. https://doi.org/10.3390/medicina60081270

AMA Style

Shalata W, Abu Jama A, Dudnik Y, Abu Saleh O, Shalata S, Tourkey L, Sheva K, Meirovitz A, Yakobson A. Adverse Events in Osimertinib Treatment for EGFR-Mutated Non-Small-Cell Lung Cancer: Unveiling Rare Life-Threatening Myelosuppression. Medicina. 2024; 60(8):1270. https://doi.org/10.3390/medicina60081270

Chicago/Turabian Style

Shalata, Walid, Ashraf Abu Jama, Yulia Dudnik, Omar Abu Saleh, Sondos Shalata, Lena Tourkey, Kim Sheva, Amichay Meirovitz, and Alexander Yakobson. 2024. "Adverse Events in Osimertinib Treatment for EGFR-Mutated Non-Small-Cell Lung Cancer: Unveiling Rare Life-Threatening Myelosuppression" Medicina 60, no. 8: 1270. https://doi.org/10.3390/medicina60081270

Article Metrics

Back to TopTop