metricas
covid
Buscar en
Clinics
Toda la web
Inicio Clinics Association of tumor mutation burden and epidermal growth factor receptor inhibi...
Journal Information
Vol. 76.
(January 2021)
Share
Share
Download PDF
More article options
Visits
956
Vol. 76.
(January 2021)
ORIGINAL ARTICLE
Open Access
Association of tumor mutation burden and epidermal growth factor receptor inhibitor history with survival in patients with metastatic stage III/IV non-small-cell lung cancer: A retrospective study
Visits
956
Yan LanI, Shuo ZhouII, Weihong FengIII, Ying QiaoI, Xueming DuIII,
Corresponding author
dudaming73@163.com

Corresponding authors.
, Fenge LiIII,IV,
Corresponding author
rosetea85@163.com

Corresponding authors.
I Department of Oncology, Chifeng Songshan Hospital, Mongolia, China
II Department of Nuclear Medicine, Provincial Clinical Hospital of Fujian Medical University, Fuzhou, China
III Department of Oncology, Tianjin Beichen Hospital, Tianjin, China
IV Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (3)
Show moreShow less
OBJECTIVES:

Lung cancer is the leading cause of cancer-related deaths worldwide. However, factors associated with the survival of patients with advanced non-small-cell lung cancer (NSCLC) who received only hospice care are largely unclear. In this study, we aimed to determine the prognostic factors correlated with survival in patients with advanced NSCLC who had undergone hospice care only.

METHODS:

A total of 102 patients with recurrent stage III/IV NSCLC after traditional treatment failure were investigated. Survival was measured from the date of enrollment to December 2019 or the time of death. Tumor tissues were collected, and DNA sequencing was performed to identify somatic mutations. Data on clinical factors of patients were collected and analyzed by univariate and multivariate analyses. Overall survival analysis was conducted using the Kaplan-Meier method.

RESULTS:

The 6-month, 1-year, and 2-year overall survival rates of the 102 patients with metastatic NSCLC were 17.65%, 3.92%, and 0.98%, respectively. The median overall survival of the 102 patients was 3.15 months. Tumor location in the peripheral lung, epidermal growth factor receptor (EGFR) inhibitor history, low tumor mutation load, adenocarcinoma, and poor performance status score were associated with prolonged survival compared with tumor location in the central lung, no EGFR inhibitor history, high tumor mutation load, squamous cell carcinoma, and good performance status score (p=0.045, p=0.003, p=0.045, p=0.021, and p=0.0003, respectively).

CONCLUSIONS:

EGFR inhibitor treatment history and tumor mutation load are risk factors for the overall survival of patients with stage III/IV NSCLC who have undergone only hospice care. These results provide a critical clinical basis for further study of nontraditional anti-tumor responses induced by EGFR inhibitors.

KEYWORDS:
Lung Cancer
Hospice Care
EGFR Inhibitor
Overall Survival
Tumor Mutation
Full Text
INTRODUCTION

Lung cancer is the leading cause of cancer-related deaths worldwide (1). Non-small-cell lung cancer (NSCLC), which accounts for approximately 85% of all lung cancer cases, is often diagnosed at a late stage and has a poor prognosis (2). Traditional treatment strategies, including surgical resection and chemotherapy, are most commonly used in lung cancer treatment. However, the survival prognosis achieved with these conventional therapies is still unsatisfactory, with a 5-year survival rate of only approximately 15% (3–4). Recently, epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) gefitinib, erlotinib, and AZD-92912 have been used to treat unresectable or recurrent lung cancer and have shown significantly improved clinical outcomes in patients with NSCLC with EGFR mutations (5–15). Nonetheless, tumors will inevitably develop drug resistance, and tumor recurrence has been observed (16–18).

Some patients who experienced disease progression, continued on conventional treatments following failures. It has been shown that continuing EGFR inhibitor treatment is beneficial in many patients even after they develop resistance to EGFR inhibitors on the basis of the hypothesis that a population of EGFR inhibitor-sensitive cells remains during disease progression, and resistant cells may be detected radiographically before widespread dissemination occurs (19). Other patients receive only hospice care. Patients who discontinue EGFR inhibitor treatment have a higher risk of symptomatic progression and increase in tumor size, which may lead to a much more rapid progression of the cancer (20–21). Despite the success of EGFR inhibitor treatment, questions on whether the benefits of continuing EGFR inhibitor treatment are temporary or long-term, how the overall survival is affected after EGFR inhibitor discontinuation, and what factors are correlated with overall survival in patients with metastatic stage III/IV NSCLC who receive hospice care remain unanswered. In this study, we aimed to determine the prognostic factors that are correlated with the survival of patients with advanced NSCLC who had received only hospice care.

SUBJECTS AND METHODSPatient selection

A total of 102 patients with stage III/IV advanced NSCLC between December 2015 and April 2019 were included in this study. The study protocol was in accordance with the ethical guidelines outlined in the Declaration of Helsinki, as revised in 2013, and was approved by the Tianjin Anti-Cancer Association and ethics committee of the Tianjin Beichen Hospital. Informed consent was obtained from all patients enrolled in this study. Patients were selected according to the following inclusion criteria: 1) stage III/IV NSCLC according to the National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology (version 3.2016), Non-Small-Cell Lung Cancer Stage Classification; 2) disease progression on standard treatments, including surgery, chemotherapy, radiotherapy, and targeted drug therapy, and the absence of an active treatment option; 3) current hospice care only; and 4) a diagnosis of NSCLC confirmed by biopsy. Patients were excluded from the present study if they 1) did not meet the abovementioned criteria; 2) continued to receive antineoplastic therapies intended at prolonging survival, such as traditional cytotoxic, targeted, or immune-based therapies, despite disease progression; or 2) had incomplete medical records. It is noteworthy that patients with pleural effusion were all confirmed as having malignant pleural effusion by thoracentesis. None of the enrolled patients received any other survival-prolonging treatments during the follow-up period. All patient data were retrospectively collected from detailed hospital medical records after clinical sample detection. The clinical and demographic characteristics of all patients are summarized in Table 1. A study flowchart is shown in Figure 1.

Table 1.

Patients' clinical and demographic characteristics at baseline.

Characteristic  Mean±SD or no. of cases (n=102)  Range 
Sex (male-female)  57-45  − 
Age (year)  65.64±11.20  28-91 
Weight (kg)  62.50±9.45  40.5-83 
Height (cm)  167.0±7.43  150-186 
Smoking history (yes-no)  71-31  − 
Pleural effusion (yes-no)  55-47  − 
Tumor size (cm)  4.34±1.64  1.4-10.1 
Tumor site (central-peripheral)  42-60  − 
CEA (ng/mL)  71.40±88.73  0.91-1005 
CA125 (U/mL)  110.03±257.67  11.47-2482.1 
CA153 (U/mL)  50.10±103.37  4.5-1016.2 
EGFR mutation (yes-no)  17-31**  − 
Previous treatments     
Surgery (yes-no)  7-95  − 
Local radiotherapy (yes-no)  89-13  − 
Chemotherapy (yes-no)  73-29  − 
EGFR inhibitor (yes-no)  17-85  − 
Lymphocyte (%)  18.01±8.68  3.3-44.5 
Brain metastases (yes-no)  11-91  − 
Bone metastases (yes-no)  35-67  − 
Tumor histology (SQ-AD)  48-54  − 
ECOG PS (0/1/2-3)  31-71  − 
**

48 of 102 performed EGFR mutation analysis.

Figure 1.

Flowchart of the study.

(0.02MB).
Hospice care

Hospice care is defined as supportive care at the end of life when life prolongation is not the primary treatment goal and disease-modifying therapies are no longer provided (22). Hospice care is particularly intended at improving the quality of life of patients through pain relief; treatment of fever and cough caused by lung tumor; and physical, psychosocial, and spiritual care. In this study, hospice care included the use of analgesics, antipyretic and cough medications, and antibiotics, with attention to psychological and spiritual aspects of care. None of the enrolled patients received antineoplastic treatments intended at improving survival, including conventional chemotherapy, radiotherapy, targeted drug therapy, or immunotherapy, during the follow-up period.

Assessment of somatic tumor cell mutation by next-generation sequencing

We followed a genotyping panel designed to detect 508 cancer-associated genes on the basis of the biobanking system and in conjunction with the clinic and pathology laboratory (23). DNA was extracted from tumor samples of 40 patients immediately after biopsy using the TIANamp Genomic DNA Kit (TIANGEN) according to the manufacturer's instructions. Then, exome libraries were constructed from the isolated DNA. Barcoded next-generation sequencing libraries were constructed (Hengjia Biotech), and exome captures were sequenced on the HiSeq X Ten System (Illumina, USA).

Serological test for carcinoembryonic antigen (CEA), carcinoma antigen 125 (CA125), and carcinoma antigen 153 (CA153) and lymphocyte percentage analysis

Serum CEA, CA125, and CA153 levels were determined by a luminescence-based method using detection kits (Access CEA, Cat. #33200; Access OV Monitor, Cat. #386357; Access BR Monitor, Cat. #387620, Beckman Coulter Inc.) and subsequently measured using the Automatic Luminescence Immunoassay Analyzer (UniCel DxI 800, Beckman Coulter). Lymphocyte percentage was tested by fluorescence-activated cell sorting using the auto hematology analyzer (BC-6800, Mindray).

Follow-up assessments

For survival analysis, all patients were followed up from the date of initial enrollment until December 2019 or until the time of death. Subsequent follow-up examinations included assessment of tumor biomarkers CEA, CA125, and CA153; peripheral blood T-cell counts; and tumor DNA sequencing of 40 patients from whom tissue samples were available.

Statistical analysis

Univariate and multivariate analyses were performed to evaluate the possible factors that were correlated with patient prognosis and survival outcomes. Survival time was defined as the duration from the date of enrollment to the date of death or December 2019. The enrollment date for each patient in the study was the date of start of hospice care only. Survival curves were plotted, and overall survival rates were estimated using the Kaplan-Meier method and compared using the log-rank test or Cox's proportional hazard model. Variables with p<0.6 on univariate analysis were entered into the multivariate model (Supplemental Table 1). The Cox regression method was used for multivariate analyses. Variables with p<0.05 were considered statistically significant. All statistical analyses were performed using the Statistical Package for Social Sciences (SPSS 16.0 for Windows; SPSS Inc., Chicago, IL) and GraphPad Prism 5.0 (USA).

Supplemental Table 1.

Multivariate analysis showed independent factors associated with the overall survival rates of 102 patients with NSCLC.

Variables  SE  p-value  Exp (β)  95% CI for Exp (β) 
Age:>67 years vs. ≤67 years  0.570  0.710  0.809  0.265 to 2.472 
Sex: female vs. male  0.865  0.446  1.933  0.355 to 10.528 
Height: >170 cm vs. ≤170 cm  0.705  0.778  0.820  0.206 to 3.261 
Tumor size: >4.35 cm vs. ≤5.35 cm  0.558  0.312  0.569  0.191 to 1.699 
CEA: >43.79 ng/mL vs. ≤43.79 ng/mL  0.566  0.030  3.424  1.130 to 10.380 
CA153: >34.39 u/mL vs. ≤34.39 u/mL  0.496  0.591  1.305  0.494 to 3.447 
EGFR inhibitor history: yes vs. no  0.933  0.794  0.783  0.126 to 4.878 
EGFR mutation: yes vs. no  0.546  0.051  2.897  0.994 to 8.441 
Lymphocyte: >19.6% vs. ≤19.6%  0.495  0.908  1.059  0.401 to 2.796 
Tumor pathology: SQ vs. AD*  0.793  0.012  7.275  1.536 to 34.445 
ECOG PS: 0/1/2 vs.0.652  0.019  4.626  1.289 to 16.602 
Tumor site: central vs. peripheral  0.907  0.035  6.796  1.148 to 40.223 
Chemotherapy cycles: ≥3 vs. <3  0.694  0.176  0.391  0.101 to 1.524 
Chemotherapy: yes vs. no  0.797  0.066  4.327  0.908 to 20.628 
Number of tumor somatic mutations: ≥3 vs. <3  0.724  0.077  3.591  0.870 to 14.829 
*

SQ, squamous cell carcinoma; AD, adenocarcinoma.

Bold values indicate significant difference.

RESULTSOverall survival analysis of all patients with advanced stage III/IV NSCLC who had undergone hospice care only

The 6-month, 1-year, and 2-year overall survival rates of the 102 patients with metastatic NSCLC were 17.65%, 3.92%, and 0.98%, respectively. The median overall survival of the 102 patients was 3.15 months (Figure 2), which was consistent with that in previous studies, which reported a median overall survival of 3-5 months (24–26). These results provide greatly important basic survival data to researchers evaluating new treatment strategies for patients with recurrent stage III/IV NSCLC who show progression on multiple conventional treatments.

Figure 2.

Overall survival (OS) curve of all 102 patients with advanced stage III/IV NSCLC. The 6-month, 1-year, and 2-year OS rates were 17.65%, 3.92%, and 0.98%, respectively. The median OS of all 102 studied patients was 3.15 months.

(0.04MB).
Correlation of EGFR inhibitor history with prolonged survival

Forty-eight of the 102 patients had undergone EGFR mutation evaluation, among whom 17 patients were found to have an EGFR mutation and EGFR inhibitor treatment history and 31 had no EGFR mutation or EGFR inhibitor treatment history. Detailed gene mutation signatures of each patient are shown in Supplemental Tables 2 and 3. The median overall survival time of patients with and without an EGFR inhibitor history were 6.13 months and 3.10 months, respectively (=0.038; 95% confidence interval [CI], 0.2998-0.9667) (Figure 3A, Table 2). This result suggests that EGFR inhibitor history was associated with a longer survival time than was no EGFR inhibitor history in patients. This leads to an extremely interesting question: Do EGFR inhibitors affect patient tumor progression and regression fate? Meanwhile, this result was based on a relatively small population of patients, and larger studies are needed to further confirm this finding.

Supplemental Table 2.

Gene mutation results of 40 patients according to 508 gene panel analysis.

Patient ID  No. of gene mutation  Gene  Base mutation  Amino acids mutation 
Pt. 12  TSC2  c.4519T>C  p.S1507P 
Pt. 153EGFR  c.2573T>G  p.L858R 
TP53  c.859G>T  p.E287* 
CDK4  c.71G>T  p.R24L 
EGFR  1.7copy number again 
Pt. 199TP53  c.337T>G  p.F113V 
RB1  c.1450_1451del  p.M484Vfs*8 
CD22  c.A139A>G  p.R47G 
PIK3C2A  c.1201C>T  p.R401C 
EPHA5  c.2767C>A  p.P923T 
NEK11  c.25A>G  p.K9E 
STAG2  c.3446A>T  p.H1149L 
EPHA5  c.688T>G  p.Y230D 
ATM  c.4966A>G  p.K1656E 
Pt. 21  ERBB2  c.338G>A  p.R113Q 
Pt. 22  EGFR  c.2573T>G  p.L858R 
Pt. 236RAD52  c.1037C>A  p.S346X 
CREBBP  c.1651C>A  p.L551I 
TEK  c.2029C>A  p.Q677K 
PIK3C3  c.718G>A  p.G240S 
MPL  c.1120A>G  p.T374A 
PIK3C2G  c.3299_3306dup  p.Y1103Vfs*15 
Pt. 252TP53  c.469G>T  p.V157F 
CDKN2A  c.250G>A  p.D84N 
Pt. 267NOTCH2  c.2548G>A  p.E850K 
APC  c.4666dup  p.T1556Nfs*3 
TP53  c.532del  p.H178Tfs*69 
MS4A1  c.161C>A  p.A54D 
KEAP1  c.1337A>T  p.E446V 
IGF1R  c.758A>G  p.H253R 
MLH3  c.2032A>G  p.N678D 
Pt. 28  EGFR  c.2235_2249del  p.745_750del 
Pt. 319PTCH1  c.C3853>T  p.Q1285X 
KMT2D  c.10636C >T  p.Q3546X 
CARD11  c.3179G>A  p.C1060Y 
EXT1  c.1813C>T  p.R605W 
TRAF7  c.694C>T  p.R232W 
DNMT3A  c.1574C>T  p.A525V 
CCND3  c.391G>A  p.A131T 
ERBB2  c.1830G>T  p.K610N 
TAF1  c.3650G>A  p.R1217H 
Pt. 352CHUK  c.464T>C  p.V155A 
PTCH1  c.2225G>A  p.R742H 
Pt. 39  TP53  c.527G>T  p.C176F 
Pt. 422KRAS  c.34G>A  p.G12S 
TP53  c.775G>T  p.D259Y 
Pt. 449ABL2  c.2948C>T  p.A983V 
BRCA2  c.8187G>T  p.K2729N 
DOCK2  c.4768A>G  p.R1590G 
ERBB2  c.380G>A  p.R127Q 
Pt. 447EGFR  c.2573T>G  p.L858R 
TP53  c.584T>C  p.I195T 
CDKN2A  c.315C>A  p.D105E 
NTRK1  c.2026C>T  p.R676C 
ALK  c.3311C>T  p.S1104F 
FGFR2  c.160C>T  p.P54S 
EGFR  1.7copy number again 
Pt. 472EGFR  c.2237_2254del  p.E746_S752delinsA 
EGFR  c.2255C>T  p.S752F 
BRCA1  c.718C>T  p.Q240X 
Pt. 505AKT1  c.607C>T  p.Q203X 
BRAF  c.1568C>T  p.P523L 
FGFR1  c.748C>T  p.R250W 
NF1  c.4998G>C  p.R1666S 
GOPC  c.1151G>A  p.R384H 
Pt. 537EGFR  c.2573T>G  p.L858R 
KMT2C  c.817delG  p.V273Wfs*82 
NFE2L3  c.1826_1829del  p.C611Tfs*22 
SMC1A  c.3103C>T  p.R1035X 
FAT3  c.12637C>T  p.R4213C 
SMAD4  c.608C>T  p.P203L 
PRKCG  c.614G>A  p.R205Q 
Pt. 633EGFR  c.2573T>G  p.L858R 
KRAS  c.35G>A  p.G12D 
TSC2  c.4594C>T  p.Q1532X 
Pt. 642STK11  c.1062C>G  p.F354L 
TP53  c.430C>T  p.Q144X 
Pt. 656NTRK1  c.865C>T  p.Q289X 
PMS2  c.80G>T  p.C27F 
ATM  c.7358G>C  p.R2453P 
SMAD4  c.608C>T  p.P203L 
TSC1  c.2696C>G  p.T899S 
PTEN  c.235G>A  p.A79T 
Pt. 66  TP53  c.524G>A  p.R175H 
Pt. 672EGFR  c.2573T>G  p.L858R 
TP53  c.430C>T  p.Q144X 
Pt. 684DDR2  c.1267C>T  p.Q423X 
MET  c.1039G>A  p.A347T 
PTCH1  c.4144C>T  p.H1382Y 
HER2  c.2579C>T  p.A860V 
Pt. 695EGFR  c.2232_2233ins  p.I744_K745insKIP VAI 
MSH3  c.1180C>T  p.Q394X 
AR  c.1369_1377del  p.G457_G459del 
PTCH1  c.1628G>A  p.R543H 
TSC2  c.2023G>A  p.A675T 
Pt. 702PIK3CA  c.1633G>A  p.E545K 
TP53  c.524G>A  p.R175H 
TP53  c.1006G>T  p.E336X 
Pt. 712EGFR  c.2156G>C  p.G719A 
TP53  c.661G>T  p.E221X 
TP53  c.745A>T  p.R249W 
Pt. 72  TP53  c.524G>A  p.R175H 
Pt. 73  EGFR  c.2237_2251delinsT AG  p.E746_T751delins VA 
Pt. 745SMARCA4  c.1189C>T  p.R397X 
DPYD  c.1850C>T  p.T617M 
XPO1  c.740T>C  p.I247T 
PRKAA1  c.1382G>A  p.R461Q 
SMC1A  c.1990C>T  p.R664W 
Pt. 752EGFR  c.2235_2249del  p.745_750del 
TP53  c.724T>A  p.C242S 
Pt. 765U2AF1  c.101C>T  p.S34F 
CD22  c.2182C>T  p.Q728X 
SETD2  c.1814A>C  p.K605T 
TNFAIP3  c.1344G>C  p.W448C 
CARD11  c.157G>A  p.D53N 
Pt. 773MLH1  c.1192C>T  p.Q398X 
AR  c.2257C>T  p.R753X 
TSC2  c.5051_5068del  p.1684_1690del 
Pt. 782EGFR  c.2575G>A  p.A859T 
TP53  c.524G>A  p.R175H 
Pt. 792NF1  c.2023G>T  p.G675X 
TP53  c.532delC  p.H178fs 
Pt. 802EGFR  c.2573T>G  p.L858R 
EGFR  c.2240T>C  p.L747S 
TP53  c.832C>T  p.P278S 
Pt. 813ERBB2  Copy number gain   
TP53  c.245_246insCACC  p.P82fs 
MET  c.3520G>T  p.V1174F 
Pt. 82  None     
Pt. 832TP53  c.536A>G  p.H179R 
TSC2  c.2023G>A  p.A675T 
Pt. 842U2AF1  c.101C>T  p.S34F 
TP53  c.994-1G>T   
Supplemental Table 3.

EGFR mutation results of eight patients who had undergone single EGFR gene analysis.

Patient ID  EGFR status 
Pt. 6  No EGFR mutation 
Pt. 11  No EGFR mutation 
Pt. 24  EGFR Exon21 p.L858R 
Pt. 27  EGFR Exon 19 c.2235_2249del p.746_750del 7.17% 
Pt. 41  No EGFR mutation 
Pt. 48  No EGFR mutation 
Pt. 60  EGFR Exon21 p.L858R 
Pt. 34  No EGFR mutation 
Figure 3.

A. Patients with an EGFR inhibitor history showed better survival than did patients without an EGFR inhibitor history. B. Patients with a higher tumor mutation load survived for shorter periods than did patients with a lower tumor mutation burden. C. Patients with a tumor in the peripheral lung lived for significantly longer than did patients with a tumor in the central lung. D. There is statistical difference in overall survival between patients with squamous cell carcinoma and those with adenocarcinoma. Survival analysis was conducted using the Kaplan-Meier method.

(0.06MB).
Table 2.

Factors associated with overall survival in 102 patients with advanced NSCLC according to univariate analysis.

Characteristic  Median or no. of cases  Median survival time (months) vs. ≥median(or left vs. right)  p-value  Chi-square  95% CI of hazard ratio 
Sex (male-female)  57-45  3.07 vs. 4.23  0.0305  4.683  1.042 to 2.309 
Age (year)  67  3.10 vs. 3.27  0.5676  0.3266  0.5887 to 1.337 
Weight (kg)  162  3.27 vs. 3.10  0.6156  0.2520  0.6066 to 1.345 
Height (cm)  170  3.97 vs. 2.92  0.1085  2.577  0.4759 to 1.077 
Smoke history (yes-no)  71-31  3.2 0 vs. 3.10  0.7461  0.1048  0.6018 to 1.439 
Pleural effusion (yes-no)  55-47  3.07 vs. 3.27  0.6862  0.1633  0.7309 to 1.610 
Tumor size (cm)  4.35  2.93 vs. 3.23  0.5509  0.3557  0.5904 to 1.325 
Tumor site (central-peripheral)  42-60  3.08 vs. 3.25  0.0451  4.013  1.009 to 2.375 
CEA (ng/mL)  43.79  3.20 vs. 3.09  0.2187  1.513  0.5124 to 1.165 
CA125 (u/mL)  65.99  2.85 vs. 3.95  0.8087  0.0586  0.7034 to 1.570 
CA153 (u/mL)  34.39  3.47 vs. 3.02  0.4449  0.5835  0.5745 to 1.276 
EGFR mutation (yes-no)  17-31*  6.13 vs. 3.10  0.0381  4.298  0.2998 to 0.9667 
Number of tumor somatic mutations  3**  4.27 vs. 2.43  0.0451  4.017  0.2365 to 0.9841 
Previous treatments           
Surgery (yes-no)  7-95  3.10 vs. 3.20  0.4324  0.6165  0.5879 to 3.459 
Local radiotherapy (yes-no)  89-13  3.20 vs. 3.10  0.7088  0.1394  0.4807 to 1.646 
Chemotherapy (yes-no)  73-29  3.20 vs. 2.93  0.5201  0.4137  0.7518 to 1.758 
EGFR inhibitor (yes-no)  17-31*  6.13 vs. 3.10  0.0381  4.298  0.2998 to 0.9667 
Chemotherapy cycles  3.60 vs. 3.10  0.3435  0.8973  0.5440 to 1.236 
Lymphocyte (%)  19.6  2.87 vs. 4.01  0.0894  2.885  0.9484 to 2.097 
Brain metastases (yes-no)  11-91  2.87 vs. 3.20  0.9912  0.0001  0.5281 to 1.880 
Bone metastases (yes-no)  35-67  3.10 vs. 3.20  0.8146  0.0550  0.6287 to 1.440 
Metastases (visceral-bone)  17-35  2.93 vs. 3.10  0.6422  0.3714  0.6540 to 2.243 
Disease stage (III-IV)  38-64  3.20 vs. 3.10  0.6973  0.2791  0.7367 to 1.701 
Tumor histology (SQ-AD)*  48-54  3.00 vs. 3.62  0.0205  5.367  0.4020 to 0.9268 
ECOG PS (0/1/2-3)  31-71  4.58 vs. 2.77  0.0003  13.05  0.3125 to 0.7082 
*

48 of 102 underwent EGFR mutation analysis;

**

40 of 102 patients underwent 508 gene panel analysis (23). SQ, squamous cell carcinoma; AD, adenocarcinoma

Higher somatic mutation load is associated with worse overall survival

We were able to collect tumor tissues from 40 of 102 patients and performed the 508 gene panel next-generation DNA sequencing analysis. A range of 0 to 9 mutations was detected in 40 patients (Supplemental Table 2). Univariate analysis of these 40 patients showed that >3 somatic mutations was associated with a shorter overall survival than with <3 somatic mutations (4.27 months vs. 2.43 months, p=0.045; 95% CI, 0.2365-0.9841) (Figure 3B, Table 2). This result implies that tumor cell mutation load impacts tumor cell growth, migration, and progression through possibly multiple unknown signaling pathways. Further mechanistic studies are necessary to better understand lung cancer progression.

Potential correlation of demographic characteristics with patient prognoses

In addition to EGFR inhibitor history and tumor somatic mutation burden, we also found that tumor location within the lung and patient performance status score were associated with survival outcome. Patients with tumors in the central lung had extended survival compared with patients with tumors in the peripheral lung (3.25 months vs. 3.08 months, p=0.045; 95% CI, 1.009-2.375) (Figure 3C, Table 2). A patient performance status score-ECOG performance status of 0-2 was correlated with better survival than with an ECOG PS of 3 (4.58 months vs. 2.77 months, p=0.0003; 95% CI, 0.3125-0.7082 ) (Table 2). Furthermore, patients with adenocarcinoma and EGFR mutations showed a longer overall survival than did patients with squamous cell carcinoma and without EGFR mutation (p=0.021 and p=0.038, respectively). Further multivariate analysis showed that tumor pathology, ECOG performance status, and tumor site were independent factors associated with the overall survival of these patients (p=0.012, p=0.019, p=0.035, respectively). Curiously, clinical factors including pleural effusion; tumor size; tumor biomarkers CEA, CA125, and CA153; chemotherapy cycles; and brain metastases did not significantly affect survival in these patients and were therefore not considered risk factors (p=0.686, p=0.5509, p=0.219, p=0.809, p=0.445, p=0.344, and p=0.991, respectively; Figure 3D, Table 2). Our findings suggest that these clinical factors may not be correlated with survival in this terminal disease stage. However, these results are based on a small population of cases and particular background of patients and need to be further confirmed with extended studies of a larger size.

DISCUSSION

Recently, much progress has been made toward advancing lung cancer treatments, such as EGFR mutation-based targeted therapies (27). However, the prognoses of late-stage lung cancer remain poor, as almost all patients eventually develop drug resistance and disease progression within one or two years (28–29). This is particularly important in Asia, as approximately 40% of patients with NSCLC carry EGFR mutations (23). What causes the difference between EGFR mutant and non-mutant tumors and how the tumor biology changes after EGFR inhibitor treatment failure remain largely unknown. Our group, along with many others, has previously shown that patients with EGFR-positive NSCLC have a worse overall survival compared with patients with EGFR-negative NSCLC (23,30–32). This result suggests that EGFR mutant tumor cells may be activated in multiple tumor proliferation and migration pathways that are associated with EGFR signaling. Therefore, blocking EGFR signaling has the potential to reverse or slow tumor progression.

Here, we report that patients with advanced stage III/IV NSCLC and an EGFR inhibitor treatment history have a significantly longer overall survival than do patients without an EGFR inhibitor treatment history (p=0.038). We have shown for the first time that EGFR inhibitor treatment may benefit even patients with NSCLC undergoing hospice care only, with a better overall survival. Another interpretation from our findings could be that patients with different EGFR statuses have distinct tumor biology. EGFR acts as a cytoplasmic kinase and mediates many/several important growth factors, signaling both extracellularly and intracellularly. We hypothesize that EGFR inhibitor treatment eliminates EGFR mutant cells while sparing non-mutant cells. Compared with EGFR mutant cells, these non-mutant cells proliferate at a much slower rate and, as a consequence, contribute to longer survival in patients previously treated with EGFR inhibitors. However, this result was based on a small sample size, and large-scale studies are needed to confirm our findings.

The correlation of survival and somatic mutation load has been reported in several different cancer types (33–35). High somatic mutation is correlated with decreased progression-free survival in multiple myeloma and tumor progression in melanoma (33–34). Interestingly, tumor mutation load is significantly correlated with the clinical outcome of anti-CTLA4 antibody and adoptive T-cell treatment in melanoma. The clinical benefit of anti-PD-1 treatment in lung cancer is also found to be strongly associated with tumor mutation load status (35). These results suggest that tumor cells with high mutation load are more aggressive in nature but are simultaneously more sensitive to immunotherapy. Consistent with these findings, we showed that patients with metastatic stage III/IV NSCLC and a higher tumor mutation load survived for shorter periods than did patients with metastatic stage III/IV NSCLC with a lower tumor mutation load (p=0.045). It is noteworthy that these patients were not under any antineoplastic treatment, and the result reflects the actual effect of tumor mutation load on overall survival.

Additionally, we analyzed other clinical factors that may be associated with the survival of patients with advanced stage III/IV NSCLC who were under hospice care only. Tumor site and performance status score were significantly correlated with patient survival, whereas tumor size; pleural effusion; tumor biomarkers CEA, CA125, and CA153; chemotherapy cycles; and brain metastases were not. Patients with tumors that initiated in or were localized to the peripheral lung lived longer than did those with tumors in the central lung. The reason for this is that tumors developing in the central lung are more likely to block the main airway and consequently lead to death. We also measured the overall survival of all patients, showing that the median overall survival was 3.15 months. The 6-month, 1-year, and 2-year overall survival rates were 17.65%, 3.92%, and 0.98%, respectively. These data establish a primary survival baseline for developing new treatment strategies in future studies.

Taken together, this study reveals that EGFR inhibitor history and tumor mutation load may influence the prognosis of patients even at the terminal disease stage. While the changes in tumor biology after EGFR inhibitor treatment and how immunogenic tumors continue to progress after treatment remain unknown, ongoing research is crucial to better understand the role of the EGFR signaling pathway and tumor mutation load in lung tumor cell fate determination and, in turn, help determine appropriate treatment modulation in patients with advanced lung cancer.

AUTHOR CONTRIBUTIONS

Lan Y was responsible for the study design. Zhou S was responsible for the data collection and analysis. Feng W was responsible for the data collection. Qiao Y was responsible for the study design and project supervision. Du X was responsible for the study design and data analysis. Li F was responsible for the manuscript writing and data analysis.

ACKNOWLEDGMENTS

This work was supported by the Tianjin Beichen Hospital.

APPENDIX

Supplemental Table 1.

Multivariate analysis showed independent factors associated with the overall survival rates of 102 patients with NSCLC.

Variables  SE  p-value  Exp (β)  95% CI for Exp (β) 
Age:>67 years vs. ≤67 years  0.570  0.710  0.809  0.265 to 2.472 
Sex: female vs. male  0.865  0.446  1.933  0.355 to 10.528 
Height: >170 cm vs. ≤170 cm  0.705  0.778  0.820  0.206 to 3.261 
Tumor size: >4.35 cm vs. ≤5.35 cm  0.558  0.312  0.569  0.191 to 1.699 
CEA: >43.79 ng/mL vs. ≤43.79 ng/mL  0.566  0.030  3.424  1.130 to 10.380 
CA153: >34.39 u/mL vs. ≤34.39 u/mL  0.496  0.591  1.305  0.494 to 3.447 
EGFR inhibitor history: yes vs. no  0.933  0.794  0.783  0.126 to 4.878 
EGFR mutation: yes vs. no  0.546  0.051  2.897  0.994 to 8.441 
Lymphocyte: >19.6% vs. ≤19.6%  0.495  0.908  1.059  0.401 to 2.796 
Tumor pathology: SQ vs. AD*  0.793  0.012  7.275  1.536 to 34.445 
ECOG PS: 0/1/2 vs.0.652  0.019  4.626  1.289 to 16.602 
Tumor site: central vs. peripheral  0.907  0.035  6.796  1.148 to 40.223 
Chemotherapy cycles: ≥3 vs. <3  0.694  0.176  0.391  0.101 to 1.524 
Chemotherapy: yes vs. no  0.797  0.066  4.327  0.908 to 20.628 
Number of tumor somatic mutations: ≥3 vs. <3  0.724  0.077  3.591  0.870 to 14.829 
*

SQ, squamous cell carcinoma; AD, adenocarcinoma.

Bold values indicate significant difference.

Supplemental Table 2.

Gene mutation results of 40 patients according to 508 gene panel analysis.

Patient ID  No. of gene mutation  Gene  Base mutation  Amino acids mutation 
Pt. 12  TSC2  c.4519T>C  p.S1507P 
Pt. 153EGFR  c.2573T>G  p.L858R 
TP53  c.859G>T  p.E287* 
CDK4  c.71G>T  p.R24L 
EGFR  1.7copy number again 
Pt. 199TP53  c.337T>G  p.F113V 
RB1  c.1450_1451del  p.M484Vfs*8 
CD22  c.A139A>G  p.R47G 
PIK3C2A  c.1201C>T  p.R401C 
EPHA5  c.2767C>A  p.P923T 
NEK11  c.25A>G  p.K9E 
STAG2  c.3446A>T  p.H1149L 
EPHA5  c.688T>G  p.Y230D 
ATM  c.4966A>G  p.K1656E 
Pt. 21  ERBB2  c.338G>A  p.R113Q 
Pt. 22  EGFR  c.2573T>G  p.L858R 
Pt. 236RAD52  c.1037C>A  p.S346X 
CREBBP  c.1651C>A  p.L551I 
TEK  c.2029C>A  p.Q677K 
PIK3C3  c.718G>A  p.G240S 
MPL  c.1120A>G  p.T374A 
PIK3C2G  c.3299_3306dup  p.Y1103Vfs*15 
Pt. 252TP53  c.469G>T  p.V157F 
CDKN2A  c.250G>A  p.D84N 
Pt. 267NOTCH2  c.2548G>A  p.E850K 
APC  c.4666dup  p.T1556Nfs*3 
TP53  c.532del  p.H178Tfs*69 
MS4A1  c.161C>A  p.A54D 
KEAP1  c.1337A>T  p.E446V 
IGF1R  c.758A>G  p.H253R 
MLH3  c.2032A>G  p.N678D 
Pt. 28  EGFR  c.2235_2249del  p.745_750del 
Pt. 319PTCH1  c.C3853>T  p.Q1285X 
KMT2D  c.10636C >T  p.Q3546X 
CARD11  c.3179G>A  p.C1060Y 
EXT1  c.1813C>T  p.R605W 
TRAF7  c.694C>T  p.R232W 
DNMT3A  c.1574C>T  p.A525V 
CCND3  c.391G>A  p.A131T 
ERBB2  c.1830G>T  p.K610N 
TAF1  c.3650G>A  p.R1217H 
Pt. 352CHUK  c.464T>C  p.V155A 
PTCH1  c.2225G>A  p.R742H 
Pt. 39  TP53  c.527G>T  p.C176F 
Pt. 422KRAS  c.34G>A  p.G12S 
TP53  c.775G>T  p.D259Y 
Pt. 449ABL2  c.2948C>T  p.A983V 
BRCA2  c.8187G>T  p.K2729N 
DOCK2  c.4768A>G  p.R1590G 
ERBB2  c.380G>A  p.R127Q 
Pt. 447EGFR  c.2573T>G  p.L858R 
TP53  c.584T>C  p.I195T 
CDKN2A  c.315C>A  p.D105E 
NTRK1  c.2026C>T  p.R676C 
ALK  c.3311C>T  p.S1104F 
FGFR2  c.160C>T  p.P54S 
EGFR  1.7copy number again 
Pt. 472EGFR  c.2237_2254del  p.E746_S752delinsA 
EGFR  c.2255C>T  p.S752F 
BRCA1  c.718C>T  p.Q240X 
Pt. 505AKT1  c.607C>T  p.Q203X 
BRAF  c.1568C>T  p.P523L 
FGFR1  c.748C>T  p.R250W 
NF1  c.4998G>C  p.R1666S 
GOPC  c.1151G>A  p.R384H 
Pt. 537EGFR  c.2573T>G  p.L858R 
KMT2C  c.817delG  p.V273Wfs*82 
NFE2L3  c.1826_1829del  p.C611Tfs*22 
SMC1A  c.3103C>T  p.R1035X 
FAT3  c.12637C>T  p.R4213C 
SMAD4  c.608C>T  p.P203L 
PRKCG  c.614G>A  p.R205Q 
Pt. 633EGFR  c.2573T>G  p.L858R 
KRAS  c.35G>A  p.G12D 
TSC2  c.4594C>T  p.Q1532X 
Pt. 642STK11  c.1062C>G  p.F354L 
TP53  c.430C>T  p.Q144X 
Pt. 656NTRK1  c.865C>T  p.Q289X 
PMS2  c.80G>T  p.C27F 
ATM  c.7358G>C  p.R2453P 
SMAD4  c.608C>T  p.P203L 
TSC1  c.2696C>G  p.T899S 
PTEN  c.235G>A  p.A79T 
Pt. 66  TP53  c.524G>A  p.R175H 
Pt. 672EGFR  c.2573T>G  p.L858R 
TP53  c.430C>T  p.Q144X 
Pt. 684DDR2  c.1267C>T  p.Q423X 
MET  c.1039G>A  p.A347T 
PTCH1  c.4144C>T  p.H1382Y 
HER2  c.2579C>T  p.A860V 
Pt. 695EGFR  c.2232_2233ins  p.I744_K745insKIP VAI 
MSH3  c.1180C>T  p.Q394X 
AR  c.1369_1377del  p.G457_G459del 
PTCH1  c.1628G>A  p.R543H 
TSC2  c.2023G>A  p.A675T 
Pt. 702PIK3CA  c.1633G>A  p.E545K 
TP53  c.524G>A  p.R175H 
TP53  c.1006G>T  p.E336X 
Pt. 712EGFR  c.2156G>C  p.G719A 
TP53  c.661G>T  p.E221X 
TP53  c.745A>T  p.R249W 
Pt. 72  TP53  c.524G>A  p.R175H 
Pt. 73  EGFR  c.2237_2251delinsT AG  p.E746_T751delins VA 
Pt. 745SMARCA4  c.1189C>T  p.R397X 
DPYD  c.1850C>T  p.T617M 
XPO1  c.740T>C  p.I247T 
PRKAA1  c.1382G>A  p.R461Q 
SMC1A  c.1990C>T  p.R664W 
Pt. 752EGFR  c.2235_2249del  p.745_750del 
TP53  c.724T>A  p.C242S 
Pt. 765U2AF1  c.101C>T  p.S34F 
CD22  c.2182C>T  p.Q728X 
SETD2  c.1814A>C  p.K605T 
TNFAIP3  c.1344G>C  p.W448C 
CARD11  c.157G>A  p.D53N 
Pt. 773MLH1  c.1192C>T  p.Q398X 
AR  c.2257C>T  p.R753X 
TSC2  c.5051_5068del  p.1684_1690del 
Pt. 782EGFR  c.2575G>A  p.A859T 
TP53  c.524G>A  p.R175H 
Pt. 792NF1  c.2023G>T  p.G675X 
TP53  c.532delC  p.H178fs 
Pt. 802EGFR  c.2573T>G  p.L858R 
EGFR  c.2240T>C  p.L747S 
TP53  c.832C>T  p.P278S 
Pt. 813ERBB2  Copy number gain   
TP53  c.245_246insCACC  p.P82fs 
MET  c.3520G>T  p.V1174F 
Pt. 82  None     
Pt. 832TP53  c.536A>G  p.H179R 
TSC2  c.2023G>A  p.A675T 
Pt. 842U2AF1  c.101C>T  p.S34F 
TP53  c.994-1G>T   
Supplemental Table 3.

EGFR mutation results of eight patients who had undergone single EGFR gene analysis.

Patient ID  EGFR status 
Pt. 6  No EGFR mutation 
Pt. 11  No EGFR mutation 
Pt. 24  EGFR Exon21 p.L858R 
Pt. 27  EGFR Exon 19 c.2235_2249del p.746_750del 7.17% 
Pt. 41  No EGFR mutation 
Pt. 48  No EGFR mutation 
Pt. 60  EGFR Exon21 p.L858R 
Pt. 34  No EGFR mutation 

REFERENCES
[1]
LA Torre , RL Siegel , A Jemal .
Lung Cancer Statistics.
[2]
RL Siegel , KD Miller , A Jemal .
Cancer statistics, 2017.
[3]
A Jemal , F Bray , MM Center , J Ferlay , E Ward , D Forman .
Global cancer statistics.
[4]
C Reddy , D Chilla , J Boltax .
Lung cancer screening: a review of available data and current guidelines.
Hosp Pract (1995), 39 (2011), pp. 107-112
[5]
R Rosell , E Carcereny , R Gervais , A Vergnenegre , B Massuti , E Felip , 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.
[6]
M Maemondo , A Inoue , K Kobayashi , S Sugawara , S Oizumi , H Isobe , et al.
Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR.
[7]
LV Sequist , JC Yang , N Yamamoto , K O’Byrne , V Hirsh , T Mok , et al.
Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations.
[8]
L Paz-Ares , EH Tan , K O’Byrne , L Zhang , V Hirsh , M Boyer , et al.
Afatinib versus gefitinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: overall survival data from the phase IIb LUX-Lung 7 trial.
[9]
YL Wu , Y Cheng , X Zhou , KH Lee , K Nakagawa , S Niho , et al.
Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial.
[10]
BJ Solomon , T Mok , DW Kim , YL Wu , K Nakagawa , T Mekhail , et al.
First-line crizotinib versus chemotherapy in ALK-positive lung cancer.
[11]
S Peters , DR Camidge , AT Shaw , S Gadgeel , JS Ahn , DW Kim , et al.
Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer.
[12]
AT Shaw , TM Kim , L Crinò , C Gridelli , K Kiura , G Liu , et al.
Ceritinib versus chemotherapy in patients with ALK-rearranged non-small-cell lung cancer previously given chemotherapy and crizotinib (ASCEND-5): a randomised, controlled, open-label, phase 3 trial.
[13]
DW Kim , M Tiseo , MJ Ahn , KL Reckamp , KH Hansen , SW Kim , et al.
Brigatinib in Patients With Crizotinib-Refractory Anaplastic Lymphoma Kinase-Positive Non-Small-Cell Lung Cancer: A Randomized, Multicenter Phase II Trial.
[14]
AT Shaw , TM Bauer , E Felip , B Besse , LP James , JS Clancy , et al.
Clinical activity and safety of PF-06463922 from a dose escalation study in patients with advanced ALK+ or ROS1+NSCLC.
J Clin Oncol, 33 (2015),
[15]
AT Shaw , SH Ou , YJ Bang , DR Camidge , BJ Solomon , R Salgia , et al.
Crizotinib in ROS1-rearranged non-small-cell lung cancer.
[16]
M Santarpia , N Gil , R Rosell .
Strategies to overcome resistance to tyrosine kinase inhibitors in non-small-cell lung cancer.
[17]
TS Mok , YL Wu , MJ Ahn , MC Garassino , HR Kim , SS Ramalingam , et al.
Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer.
[18]
Q Ou , X Wu , H Bao , X Tong , X Wang , X Zhang , et al.
Investigating novel resistance mechanisms to third generation EGFR TKI osimertinib in non-small cell lung cancer patients using next generation sequencing.
[19]
K Becker , Y Xu .
Management of tyrosine kinase inhibitor resistance in lung cancer with EGFR mutation.
[20]
GJ Riely , MG Kris , B Zhao , T Akhurst , DT Milton , E Moore , et al.
Prospective assessment of discontinuation and reinitiation of erlotinib or gefitinib in patients with acquired resistance to erlotinib or gefitinib followed by the addition of everolimus.
[21]
JE Chaft , GR Oxnard , CS Sima , MG Kris , VA Miller , GJ Riely .
Disease flare after tyrosine kinase inhibitor discontinuation in patients with EGFR-mutant lung cancer and acquired resistance to erlotinib or gefitinib: implications for clinical trial design.
[22]
P Bhattacharya , SK Dessain , TL Evans .
Palliative Care in Lung Cancer: When to Start.
[23]
F Li , X Du , H Zhang , T Ju , C Chen , Q Qu , et al.
Next-generation sequencing of Chinese stage IV lung cancer patients reveals an association between EGFR mutation status and survival outcome.
[24]
A Vergnenàgre , S Hominal , AE Tchalla , H Bérard , I Monnet , G Fraboulet , et al.
Assessment of palliative care for advanced non-small-cell lung cancer in France: a prospective observational multicenter study (GFPC 0804 study).
[25]
KT Duggan , S Hildebrand Duffus , RB D’Agostino Jr , WJ Petty , NP Streer , RC Stephenson .
The Impact of Hospice Services in the Care of Patients with Advanced Stage Nonsmall Cell Lung Cancer.
[26]
KJ Duggan , J Wiltshire , R Strutt , MM Boxer , A Berthelsen , J Descallar , et al.
Palliative care and psychosocial care in metastatic non-small cell lung cancer: factors affecting utilisation of services and impact on patient survival.
[27]
PM Forde , DS Ettinger .
Targeted therapy for non-small-cell lung cancer: past, present and future.
Expert Rev Anticancer Ther, 13 (2013), pp. 745-758
[28]
D Jackman , W Pao , GJ Riely , JA Engelman , MG Kris , PA Jänne , et al.
Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer.
[29]
DR Camidge , W Pao , LV Sequist .
Acquired resistance to TKIs in solid tumours: learning from lung cancer.
[30]
GV Scagliotti , G Selvaggi , S Novello , FR Hirsch .
The biology of epidermal growth factor receptor in lung cancer.
[31]
D Veale , N Kerr , GJ Gibson , PJ Kelly , AL Harris .
The relationship of quantitative epidermal growth factor receptor expression in non-small cell lung cancer to long term survival.
[32]
M Volm , W Rittgen , P Drings .
Prognostic value of ERBB-1, VEGF, cyclin A, FOS, JUN and MYC in patients with squamous cell lung carcinomas.
[33]
A Miller , Y Asmann , L Cattaneo , E Braggio , J Keats , D Auclair , et al.
High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma.
[34]
J Tiffen , S Wilson , SJ Gallagher , P Hersey , FV Filipp .
Somatic Copy Number Amplification and Hyperactivating Somatic Mutations of EZH2 Correlate With DNA Methylation and Drive Epigenetic Silencing of Genes Involved in Tumor Suppression and Immune Responses in Melanoma.
[35]
J Roszik , LE Haydu , KR Hess , J Oba , AY Joon , AE Siroy , et al.
Novel algorithmic approach predicts tumor mutation load and correlates with immunotherapy clinical outcomes using a defined gene mutation set.

No potential conflict of interest was reported.

Copyright © 2021. CLINICS
Article options
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos