Skip to main content
Lung India : Official Organ of Indian Chest Society logoLink to Lung India : Official Organ of Indian Chest Society
. 2022 Dec 29;40(1):48–58. doi: 10.4103/lungindia.lungindia_144_22

Navigating patient journey in early diagnosis of lung cancer in India

Bivas Biswas 1,, Deepak Talwar 2, Priti Meshram 3, Pramod K Julka 4, Anurag Mehta 5, SP Somashekhar 6, Srinivas Chilukuri 7, Abhishek Bansal 8
PMCID: PMC9894269  PMID: 36695259

ABSTRACT

Lung cancer (LC) is one of the leading causes of cancer deaths worldwide. In India, the incidence of LC is increasing rapidly, and a majority of the patients are diagnosed at advanced stages of the disease when treatment is less likely to be effective. Recent therapeutic developments have significantly improved survival outcomes in patients with LC. Prompt specialist referral remains critical for early diagnosis for improved patient survival. In the Indian scenario, distinguishing LC from benign and endemic medical conditions such as tuberculosis can pose a challenge. Hence, awareness regarding the red flags—signs and symptoms that warrant further investigations and referral—is vital. This review is an effort toward encouraging general physicians to maintain a high index of clinical suspicion for those at risk of developing LC and assisting them in refering patients with concerning symptoms to specialists or multidisciplinary teams as early as possible.

KEY WORDS: Early diagnosis, general practice, incidental pulmonary nodule, lung cancer, patient navigation, red flags, referral pathway, screening, policy consideration

INTRODUCTION

Lung cancer (LC) is the second most common cancer globally with 2,206,771 new cases and the leading cause of cancer-related deaths with 1,796,144 deaths in 2020.[1] Almost 3.3% of the world’s new LC cases and 3.7% of LC-related deaths occurred in India in 2020.[1] LC has demonstrated an increasing trend in Indian women, from 7.9% in 2008 to 27.2% in 2018.[2] It is the second leading cause of cancer mortality in men in India, whereas in women it ranks fifth.[1]

In India, 73%−93% of men and 23%−50% of women were found to be smokers at the time of diagnosis of LC[2]; approximately 59% of LC deaths in men and 15% of LC deaths in women were attributed to smoking.[3] Prevalence of LC is also high among younger patients (13.8%) and never-smokers (23.8%), and this could be possibly due to increased indoor air pollution and increased prevalence of oncogenic driver mutations, including epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements.[2]

LC is histologically classified into small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). NSCLC accounts for approximately 92% of all LC cases with adenocarcinoma (43.8%) being the predominant histological type, followed by squamous cell carcinoma (SqCC) (26.2%), large cell (2.1%), and others (8.3%).[4] In India, the prevalence of adenocarcinoma has increased by 26.4%, SCLC by 5.2%, and NSCLC-not otherwise specified has decreased by 27.8%, over a period of 10 years.[2] However, in some parts of north and east India, both adenocarcinoma and SqCC are equally predominant (36.4% each).[5]

The 5-year survival rate remains poor (9.6%) in India as opposed to Europe (<20%) and the United States (US) (15%−19%), according to a global study conducted in 67 countries.[6] An ambidirectional feasibility study from north India reported a 5-year survival rate of 2%.[7]

In India, 80% of the patients with LC consult their general practitioner (GP) or primary care physician (PCP) despite severe respiratory symptoms.[8] Timely diagnosis and treatment remain critical for survival and improved prognosis of patients with LC. However, there is an unacceptable lag of up to 6 months from symptom onset to initiation of treatment compared with studies from Western countries.[9] The reasons for the delay may be multifactorial, such as accessibility to health care, patient awareness of the disease, and aggressiveness of the diagnostic approach. Most of these factors are modifiable and can have definite implications on patient survival.

This review is an effort towards encouraging physicians to maintain a high index of clinical suspicion for those at risk of LC and assisting them in referring patients with concerning symptoms to specialists or a multidisciplinary team (MDT) as early as possible for better patient outcomes.

UNPRECEDENTED SURVIVAL BENEFITS OF LUNG CANCER

Survival of LC can be improved by early diagnosis with multimodality management, and in advanced unresectable and metastatic disease by personalised treatment using precision medicine and novel therapies. Early stages of LC have a better prognosis; early diagnosis via screening programmes could be an effective approach for reducing LC mortality. This has been proven unequivocally in asymptomatic patients in the National Lung Screening Trial.[10]

The 5-year survival rate reported in stage I disease is up to 92% and in stage II is up to 60% compared with 10% in stage IV disease.[11] However, in India, only 3.5%−7.2% of patients are diagnosed at an early stage[2,5,12,13]; the majority (90%) are diagnosed at an advanced stage of the disease.[2,5] Early diagnosis of LC may allow surgical resection, the most effective treatment, with a 5-year survival rate ranging from 60%−80% for stage I and 30%−50% for stage II NSCLC.[14] In the US, with wider availability and adoption of modern imaging modalities and invasive mediastinal staging, an improvement in LC outcomes has been observed.[15]

Early-stage LC is generally treated with surgery (lobectomy, bilobectomy, pneumonectomy), with or without adjuvant chemotherapy and/or radiotherapy. In the last few years, treatment-related morbidity and mortality have decreased with surgeries like video-assisted thoracic surgery (VATS),[16] uniportal VATS,[17] awake surgery,[18] VATS without tracheal intubation,[19] and robotic VATS.[20] Adjuvant chemotherapeutic regimens have also been shown to increase survival in resected patients with stage II and stage III disease.[21] However, stereotactic body radiotherapy has emerged as one of the most significant advances in modern radiotherapy for early-stage inoperable NSCLC and those refusing surgery, since it can deliver extremely precise radiation to very high doses in a few fractions.[22] Particularly, in appropriately selected patients, it has demonstrated comparable local control rate and 5-year survival rates to surgery in stage I NSCLC.[23,24]

More recently, the biomarker-driven treatment—specifically tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs) have substantially improved outcomes in NSCLC with data for the same being largely available from their use in advanced unresectable or metastatic disease.[15,25-27] Patients with oncogenic driver alteration -positive LC can now be treated with ease of oral treatment options with good efficacy, tolerability, and proven overall survival benefit.[27] In those without targetable oncogenic driver alterations, immunotherapy has the potential to be an effective treatment option when used either singly (high PD-L1 expression) or in combination with chemotherapy (low or absent PD-L1 expression).

CHALLENGES OF EARLY DIAGNOSIS OF LUNG CANCER

LC poses a major diagnostic challenge for PCPs, particularly in its early stages due to non-specific symptoms such as cough, chest pain, weight loss, anorexia, and breathlessness with low predictive value for the diagnosis of cancer[28,29]; only about 22% of patients with LC initially present with haemoptysis.[30]

In India, the high endemicity of tuberculosis (TB) and the overlap of its symptomatology and radiological features with LC have created a scenario where a significant number of patients (up to 42.5%) have been misdiagnosed with TB, due to a low index of suspicion toward LC.[4,8,3135] Anti-TB treatment is empirically commenced for sputum-acid-fast bacillus–negative patients with lung shadows that do not resolve with antibiotic treatment.[31,32] This could lead to a substantial delay in diagnosis of LC, which in turn may lead to stage progression and higher mortality[4,8,9,3135] [Table 1].

Table 1.

Diagnostic delays in lung cancer in India[4,8,9,31-35]

Study Type of study Place of study Period of study No. of patients (n) Stage of disease at diagnosis Diagnosis delay from presentation of symptoms (months) % of patients misdiagnosed as TB
Ramachandran et al.[8] Prospective Tamil Nadu Nov 2006 to May 2007 96 Stage IIIB and IV 5.8 29.2
Chandra et al.[9] Retrospective New Delhi Jan 2002 to Dec 2008 165 Stage IIIB and IV 4.8 17§
Noronha et al.[4] Prospective Mumbai 2008 to 2009 489 Stage I to IV 4 14.1
Shanthilal[31] Retrospective Karnataka 2011 to 2016 133 Stage III and IV 1.9* 17*
Singh et al.[32] Prospective New Delhi Nov 2007 to Nov 2008 70 - - 20
Gupta et al.[33] Prospective Jammu 2 years (year unspecified) 170 - 3 42.5
Dubey et al.[34] Prospective Madhya Pradesh 2012 to 2013 47 Stage IIIB, IV 5.7 23.4
Vashistha et al.[35] Retrospective New Delhi 2008 to 2016 1370 Stage I to IV 3.6 40*

ATT: Antituberculosis treatment; TB: Tuberculosis; *: Patients received ATT; -: Not available

Other challenges at the primary care level may include lag time from symptom onset to the first visit to the PCP, delay due to investigation and symptomatic treatment, and the diagnostic procedure to establish the cancer diagnosis apart from the non-availability of advanced diagnostic procedures and specialists.[31] Low awareness of LC symptoms,[13] financial constraints and inadequate or inaccessible healthcare facilities are some of the factors shown to modify the healthcare-seeking behaviour at the grass-roots level.[8,35]

In some cases, LC develops against a background of chronic respiratory disease and symptoms of chronic cough, typically in patients who smoke, hindering and delaying the diagnosis of LC. Anecdotal evidence points to chronic obstructive pulmonary disease (COPD)[36] and past history of TB[37] being risk factors for the development of LC.

REFERRAL PATHWAY IN SUSPECTED LUNG CANCER

Current referral pattern

In several countries, a rapid referral pattern has been introduced between the primary care and specialist clinics for patients with suspected LC[38]; but in India, no such referral pattern exists. In India, although patients may have access to a specialist, most patients fail to seek the services of a specialist directly,[8,33] and a large number of patients on average have at least two GP consultations before presenting to the pulmonologist.[8]

As per recent evidence, around 15% of the patients in India consulted pulmonologists directly at the first visit without referral and 45.3% of patients consulted general medicine specialists and 27% of the physicians referred the patients to speciality centres for evaluation.[8]

Considering the current referral and the low index of suspicion for LC among GPs, streamlining referral pathways and services is imperative to improve LC care in India.[39]

Optimising the referral pathway

Early diagnosis of LC relies mainly on prompt patient presentation and timely referral of patients with symptoms suggestive of LC by GPs to a specialist.[8,31]

Risk factors

Smoking is a well-established principal risk factor for the development of LC.[2] Anecdotal evidence points to environmental factors such as exposure to second-hand smoke, radon, emissions from combustion of solid fuels (wood, charcoal, crop residues or dung), cooking fumes, environmental particulate matter, and occupational exposures as risk factors for the development of LC in never-smokers.[40]

Medical history

In addition to a family history of LC, the previous diagnosis of other cancers, history of COPD, TB and chronic bronchitis are found to be associated with long-term risk of LC.[36,37,41]

Overlapping symptoms

In several cases, symptoms of LC may be confounded by high levels of comorbidities. Patients with LC commonly experience multiple and synchronous symptoms [Supplementary Table S1].[9,30,33,34,42] Symptoms such as cough, dyspnoea, haemoptysis, loss of appetite, weight loss, fatigue, thoracic pain, and hoarseness of voice are the warning signs of LC, especially among high-risk individuals [Supplementary Table S2].[42,43]

Red flags

Red flags are warning signs and symptoms that suggest a potentially serious underlying disease. Symptoms such as haemoptysis, unintentional weight loss, persistent cough lasting longer than 6 weeks, and clubbing of fingers are red flag symptoms and signs that require investigation and the patient’s prompt referral to a specialist.[42,43]

Individual red flag symptoms without taking other risk factors into account are likely to be inconsequential. Hence, the diagnostic approach to LC should focus on clinical history, classic symptoms,[42] symptom severity, or lack of response to treatment.

Action on red flags

Typically, an immediate chest Xray (CXR) is suggested if two or more unexplained symptoms or any one of the red flags are present for more than 2 − 3 weeks. For patients with unresolved red flag symptoms, even in the presence of a clinical or microbiological diagnosis of TB, a chest computed tomography (CT) scan is suggested to detect possible underlying malignancy. Red flag symptoms suggestive of LC present for more than 3 weeks and CXR findings, either inconclusive or indicative of LC, should always lead to a referral to a specialist.[44] If a patient presents with typical features of LC, an immediate referral is indicated rather than further investigation at the primary care level [Figure 1].

Figure 1.

Figure 1

Referral pattern in lung cancer. AFB, Acid-fast bacillus; AI, Artificial intelligence; ATT, Anti-tuberculosis treatment; CT, Computed tomography; CXR, Chest X-ray; Genxpert, Cartridge based nucleic acid amplification test; PCP, Primary care physician; TB, Tuberculosis. Inline graphic: Differential diagnosis; Inline graphic: Actionable: Inline graphic: Expert opinion; Inline graphic: opportunity; Inline graphic: Alternate action pathway; Inline graphic: referral. *malignancy, if detected at any step, should be referred to MDT for further action

MULTIDISCIPLINARY APPROACH FOR LUNG CANCER

The concept of a MDT is widely accepted as the gold standard of cancer care delivery across the world.[45,46] Studies evidenced improvements in the accuracy of staging, surgery referrals, the time interval from diagnosis to treatment and treatment receipt and clinical outcomes in LC through MDT care.[45,47,48]

The MDT panel for LC care generally comprises of a pulmonologist, medical oncologist, radiologist, thoracic surgeon, nurse, etc., and each member is assigned with clear roles and responsibilities towards the management of the patient.[48] The composition of the standard MDT panel along with the roles and responsibilities of each specialist is illustrated in Figure 2.[48] The GP can be a vital link between the patient and the MDT, leading to better coordination of referral, diagnosis and staging, and earlier assessment by oncologists and thoracic surgeons.[48] MDT approach helps to bring experts together to deliver the most appropriate evidence-based treatment options to individual patients.[49]

Figure 2.

Figure 2

Multidisciplinary team care of patient with lung cancer

Pulmonologists play a leading role in the LC MDT care in the frontline along with medical oncologists, as they are the first point of referral for patients with suspicion of LC. In early-stage LC, the involvement of a pulmonologist[50] and collaboration between oncologists and surgeons[51] are associated with increased surgical resection rates and decreased mortality rates without increased cost among patients with cancer. Also, experienced surgeons or those working in high-volume centres and who attended regular MDT meetings are more likely to work closely with treating physicians in the context of decision-making regarding adjuvant therapies for patients following surgical treatment.[52] With technical advancements in imaging techniques and tissue diagnosis, interventional pulmonologists and interventional radiologists are valuable and integral members of the MDT.[53]

ROLE OF INTERVENTIONAL RADIOLOGY OR PULMONOLOGY FOR EARLY DIAGNOSIS OF LUNG CANCER

Imaging techniques for nodule or opacity detection

Nodule characteristics (size, density, conspicuity, morphology, location) and growth rate are the cardinal parameters for the probability of identifying LC when it presents as an incidental pulmonary nodule (IPN).[54] The risk of malignancy increases with an increase in nodule size. The risk is 0.2% for a pulmonary nodule (PN) of ≤3 mm, 0.9% for PN of 4 − 7 mm, 18% for PN of 8 − 20 mm, and 50% for PN of >20 mm.[55] The risk of malignancy was 83% in PN with irregular edges.[56] Other characteristics that are associated with a higher probability of malignancy are solitary PNs located in the upper lobes, PN with spiculated or lobulated margins (88%–94% or 58%, respectively),[55] eccentric or punctated lesions[55] pure ground glass (GG) opacities (59%–73%)[56] and an increased hazy lung attenuation on the faint nodular area.[55]

The Fleischner Society guideline recommends assessing patient risk factors and nodule characteristics such as size, density, multiplicity, morphology, and growth for the management of IPN.[54] Supplementary Table S3 presents a summary of clinical guidelines for the evaluation and management of solitary IPNs.[54,57,58]

Although low-dose CT (LDCT) is preferred for LC screening programs, CXR could still provide value in the screening and diagnosis of LC. Worldwide, CXR remains the most utilised diagnostic imaging procedure for suspected LC in primary care, with reported sensitivity ranging 77%–80% for the diagnosis of symptomatic LC[59]; incidental diagnosis of LC with CXR has also been reported.[60] The sensitivity of CXR may vary according to the tumour’s size.[59] Hence, GPs should further consider an investigation, if necessary, in patients with persistent symptoms when CXR is negative. Moreover, PNs of ≤10 mm,[61] PNs in upper lobes, particularly the right lobe, and peripheral zones may be missed on CXRs, as they may be obscured by bony structures.[62,63] CXRs may not be sensitive enough to detect calcification and PN in apical (72%) or posterior segment (60%) and hilar zones.[62] Recommendations for reducing observer errors in LC identification via CXR are presented in Box 1.

Box 1.

Steps for reducing observer errors in lung cancer identification on chest X-ray[62]

Develop a specific scan path on the CXR, which covers all the lung zones symmetrically to avoid missing any zones
Always check the blind zones (apices, hila, retro-cardiac and sub-diaphragmatic spaces) and the mediastinal lines and stripes carefully
Use lateral projection, if required
Consider inverse-intensity image as an additional tool to increase confidence in identifying lesions
Compare the most recent CXR with previous CXR if available and depending on the time interval, nodule stability over a prolonged duration abolishes the need for any further action

CXR: Chest X-ray

A CT scan is more likely to show lung tumours than a routine CXR. Chest CT scan can detect small-sized PN (1–2 mm), and it can also provide specific information about the location, density, and edge characteristics of the PN.[64] However, it was recently reported that CT scans show similar GG opacities for coronavirus disease 2019 (COVID-19) and early LC, but with independent features. Hence, radiological features should be combined with epidemiological history, laboratory tests, pathological results, and short-term CT reexamination to aid differential diagnosis.[65] Contrast-enhanced CT (CECT) scan was found useful for the diagnosis of GG PNs.[66] Positron-emission tomography CT (PET-CT) scan demonstrated high accuracy in characterising PN (at least 8 mm) detected in LC with LDCT.[67] The accuracy and specificity are superior to CT scan and have less inter- and intra-observer variation.[67] In developing countries like India, owing to infectious conditions and TB, PET-CT may have a high false-positive rate and reduced specificity in characterising PN, limiting its use.[68,69] Hence, the GP must be aware of this limitation in the workup of PN, and differential diagnosis must be considered before further management decisions are taken.

Optimising tissue diagnosis

Obtaining tissue to ascertain the aetiology of a PN and establishing the diagnosis may be necessary in high-risk and intermediate-risk patients.[55] The optimal choice of technique to obtain tissue specimen depends on the type, location, and size of the lesion, comorbidities, and risk-benefit assessment of each potential strategy.

Flexible bronchoscopy is often used for the localisation of central lesions and transthoracic sampling for peripheral lesions. Over the past decade, although bronchoscopy services have witnessed their extensive use in India, they are being used only in major metropolitan cities.[70]

Different bronchoscopic techniques such as endobronchial ultrasound (EBUS), radial EBUS, electromagnetic navigation bronchoscopy, virtual 2D and 3D navigation bronchoscopy (such as Lung Point and Archimedes), and ultrathin bronchoscopy are available for the sampling of peripheral lesions. EBUS is most commonly offered by pulmonologists in India. A transthoracic biopsy is usually performed under ultrasonogram or CT by interventional radiologists. However, at present in India, less than 1% of healthcare facilities have a separate setup for interventional radiology.[71] For locating central lesions, either of the modalities is used depending on the available expertise and patient-specific factors. More recently, PET-guided biopsy has evolved as a promising technique for transthoracic sampling. It has demonstrated a 100% yield for evaluation of thoracic lesions in a patient with previous invasive sampling with inconclusive biopsy results.[72]

Most patients with the resectable disease typically undergo mediastinal staging, particularly among patients with some evidence of nodal involvement on imaging. Especially, in countries like India, where granulomatous diseases are endemic, establishing histopathological evidence of nodal involvement is particularly relevant. In India, only a few centres prefer performing mediastinal staging in all patients with resectable disease, regardless of a negative PET-CT.[73]

Mediastinoscopy is a popularly accepted diagnostic modality for mediastinal staging since it provides a larger amount of tissue for further analysis compared to endosonographic procedures. The literature has evidenced a similar yield and a lower complication rate for endoscopic procedures and mediastinoscopy. However, a higher false-negative rate has been reported for the endosonographic procedures.[74]

In recent times, peripheral blood sampling, a non-invasive way of biomarker testing is gaining importance in cancer detection because of its ease of use. Studies have reported the utility of circulating microRNAs,[75] circulating tumour DNA[76] and circulating tumour cells[77] in noninvasive diagnosis of early LC. However, active research is ongoing on the utility of these methods for diagnosing LC.

ROLE OF BIOMARKERS IN EARLY DIAGNOSIS OF LUNG CANCER

Serum biomarkers

Evaluation of tumour biomarkers in serum at an early stage of the disease has become an area of interest for many clinicians as they are minimally invasive. At the primary level, it could be useful for differentiating patients with overlapping features of TB and LC.[78] Abnormal marker levels can be considered as a criterion for the referral of patients with suspicion of LC for further evaluation to specialty centres. Mehta et al.[78] demonstrated a measurement of five tumour biomarker levels—carcinoembryonic antigen, SqCC-associated antigen, cytokeratin fragment 21-1, neuron specific-enolase, and pro-gastrinre-leasing peptide—to curtail the ambiguity of the diagnosis of LC.

Serum biomarkers are still an area of active research and cannot replace tissue for making a diagnosis of LC at present.

Molecular biomarkers

In lung adenocarcinomas, recognisable genetic driver alterations were found in 64% of the cases and their detection has implications on diagnosis, prognosis, and the use of targeted therapy.[79] A panel of molecular markers like EGFR, ALK, HER2, BRAF, ROS1, RET, and MET has been studied in LC.[80] The National Comprehensive Cancer Network® (NCCN®) currently recommends the actionable biomarkers such as EGFR (exon 19 deletions or exon 21 L858R), ALK, KRAS, NTRK1/2/3, ROS1, RET, MET exon 14 skipping and BRAF V600E testing for patients with metastatic non-squamous NSCLC before making therapy decisions, if clinically feasible.[81] However, molecular testing for these mutations or rearrangements can be considered in patients with metastatic SqCC if there is clinical suspicion of an adenocarcinoma component.[82] Programmed death-ligand 1 (PD-L1) expression and tumour mutational burden (TMB) has gained relevance as biomarkers in NSCLC, especially to evaluate clinical response to ICIs.[83] The NCCN recommends upfront PDL1 testing in newly diagnosed patients with metastatic NSCLC before commencing their treatment.[81]

In India, the prevalence of the two commonest oncogenic driver alterations, namely, EGFR mutations[2,8486] is approximately 25.3%–30% and of ALK rearrangements[2,85] is approximately 10%–11.5%, and 33.6% have PD-L1 expression.[87] The national guidelines also recommend testing for EGFR, ALK, ROS1 rearrangements, and PD-L1 overexpression for all patients with NSCLC in the frontline to provide improved treatment opportunities, including targeted therapy.[83]

SCREENING FOR LUNG CANCER IN INDIA AND POLICY CONSIDERATION

International guidelines recommend annual screening for LC with LDCT targeting high-risk patients (current or past smokers aged 50-80 years with a smoking history of 20 packyears).[88] In the National Lung Screening Trial[10] and NELSON trial,[89] about 56%−68% of the patients were detected at stage I or II. Consistently, in a study[90] 70% − 86% of the patients were diagnosed with stage I or II, which indicated that LDCT screening in community settings may significantly help in the early diagnosis of LC.[91] Besides, International Early LC Action Program results have shown a 10-year survival rate of 88% in patients with stage I disease, which was identified during screening.[92]

In many of the developing countries like Japan and Taiwan, national screening programmes are in place for early diagnosis of LC.[40] In 2016, the Ministry of Health and Family Welfare published an operational framework for the first national cancer screening programme in India.[93] According to a published framework, there will be mandatory screening for breast, oral and cervical cancer in 100 districts of India before the programme expands to other areas, targeting people over the age of 30 years.[93] However, despite the high LC incidence, no such organised national screening programmes exist in India. This may be attributed to a high prevalence of TB, poor infrastructure, logistic constraints, reluctance for screening among the high-risk population, and concerns regarding high false-positive rates.[78] In India, a policy change is an unmet need of the hour to include LC under national screening programmes along with cervical, breast, and oral cancer to propagate better outcomes for patients with LC. It could be implemented by identifying a feasible mechanism, high-risk individuals, appropriate screening tests and referral patterns, and diagnostic and therapeutic algorithms by conducting pilot studies to initiate a population-based lung screening programme.

Notably, because of the high prevalence of EGFR mutations and ALK/ROS1 rearrangements and gradually improving access to novel TKIs in the country at considerably lower costs, the Indian Council of Medical Research (ICMR) has launched a nationwide Advanced Molecular Oncology Diagnostic Services project to provide biomarker testing for LC free of cost throughout the country.[85]

ROLE OF ARTIFICIAL INTELLIGENCE

In about 90% of the cases, misdiagnosis of LC occurs on CXR.[62] Despite advancements in technology, lesions can also be overlooked due to observer errors.[62] Automated detection techniques may be a valuable tool for automatic and precise identification of possible lesions or IPNs on CXR or CT scans and thus reduce diagnostic errors.[62]

Over the last decade, with its superior ability to recognise and quantify complex patterns in images, artificial intelligence (AI) demonstrated increased diagnostic accuracy and decreased false-positive rate with automatic precise identification of possible lesions or IPNs on radiographs of the lung in CXR or CT scans captured during planned screening programmes.[94,95] A diagnostic performance of an AI for detecting LC reported an overall sensitivity of 64% and specificity of 97% for cancer-positive CXR.[96] The application of AI in imaging diagnostics aid clinicians in the interpretation of CXR or CT images and creates an opportunity for incidental identification of suspected or known LC at earlier stages. Table 2 shows the role of AI in LC diagnosis through screening and incidental identification of suspected LC.[9498]

Table 2.

Latest studies elucidating the role of artificial intelligence for nodule classification in screening, incidental identification of known or suspected lung cancer[94-98]

Study Imaging modality Study objective Study criteria Study findings Implication
Nam et al.[94] CXR Detection of malignant PN on chest radiographs A DLAD algorithm developed using 43,292 chest radiographs labelled and annotated by board-certified radiologists Algorithmic performance (radiographic classification and nodule detection) validated by one internal and four external datasets AI vs radiologist
 Specificity: 95.2% vs NA
 Sensitivity: 80.7% vs 70.4%
 Rate of false-positive findings per image: 0.30 vs 0.25
The AI algorithm, DLAD, outperformed physicians in radiograph classification and nodule detection performance for malignant PN and enhanced physicians’ performances when used as a second reader
Lee et al.[96] CXR Validate DL algorithm for LC detection in a screening population Retrospective validation of DL algorithm for LC screening detection on chest radiographs in a health screening population Validation test cohort: 10,285 radiographs Screening cohort: 10,0525 radiographs Validation results DL vs Radiologist Accuracy: 97% vs 100%
 Sensitivity: 64% vs 43%
 Specificity: 97%vs 100%
 FPR: 3.1% vs 0.3%
 Health screening results AI classification of cancer-positive CXR
 Sensitivity: 40%
 Specificity: 97%
 AI detection of visible LC
 Sensitivity: 83%
 Specificity: 97% AI detection of clearly visible LC
 Sensitivity: 100%
 Specificity: 97%
AI algorithm-DL detected LC nodules on chest radiograph with a performance comparable to that of radiologists, which will be helpful for radiologists in healthy populations with a low prevalence of LC
Liu et al.[97] CT scan Detection of LC nodules in the chest CT Five thousand 5 mm and 1 mm chest CT films of T1 stage LC patients were used to train an AI algorithm. 500 thick chest CT films of T1 stage LC patients were tested by AI algorithm, and the
 Sensitivity and
 Specificity were compared with manual film reading
AI to read 500 cases of 5 mm chest CT:
 Sensitivity: 95.20%
 Specificity: 93.20% Kappa value: 0.926,1 AI to read 500 cases of 1 mm chest CT:
 Sensitivity: 96.40%
 Specificity: 95.60%
 Kappa value: 0.938,6
The detection rates of AI and manual reading were similar for 1 mm CT sets, with no significant difference. Sensitivity of AI for 5 mm CT sets, was better than manual reading, but the number of false positives increased, and the Specificity was slightly worse. AI to automatically learn early LC chest CT images can achieve high Sensitivity and Specificity in early LC recognition and can assist doctors in diagnosis
Zhang et al.[95] CT scan Detect and classify PN derived from clinical CT images Images obtained during screening from LUNA16 and Kaggle datasets were used to pretrain the AI-CNN model CT images from four hospitals in China were used for training and validating the algorithm Data from 50 patients who underwent surgical resection and had preoperative CT were prospectively collected for final assessment of the algorithm Assessment of AI algorithm in 50-image evaluation set vs manual reading
 Accuracy: 92.0% vs 79.6%
 Sensitivity: 96.0% vs 81.3%
 Specificity: 88.0% vs 77.9%
AI compared with manual assessments exhibited significantly better performance with high Sensitivity and Specificity in detecting and classifying PN
Cui et al.[98] CT scan Identifying IPN in LDCT screening as part of routine healthcare 64,168 cases were used to retrospectively investigate the prevalence of non-calcified PNs in China by DL algorithm All CT images were automatically analysed by the DL algorithm at first. Then a junior radiologist checked the result given by the DL algorithm and revised the results when necessary Finally, an experienced radiologist confirmed the final decision and issued the diagnostic reports AI vs Radiologist
 Performance (AUC): 0.86 vs 0.73
 Sensitivity: 73% vs 83%
 Specificity 85% vs 64%
AI had better identification Sensitivity and performance than radiologists, and was highly consistent with expert radiologists in terms of PN identification, regardless of nodule size With good performance, fast processing and efficiency, AI may serve as a radiologist’s assistant

AI: Artificial intelligence; ANN: Artificial neural network; AUC: Area under curve; CNN: Convolutional neural network; CT: Computed tomography; CXR: Chest X-ray; DL: Deep learning; DLAD: Deep learning-based automatic detection; FPR: Falsepositive rate; IPN: Incidental pulmonary nodule; LC: Lung cancer; LDCT: Low-dose computed tomography; LUNA16: Lung nodule analysis 2016 challenge; NA: Not applicable; PN: Pulmonary nodule; SPN: Solitary pulmonary nodule

Thus, AI can assess CXR and CT scans for IPN in diverse settings, including primary care, acting as a facilitator and timely referral for further evaluation. Besides, AI application with CXR could be effective and economical for screening and diagnosis of LC where there is a dearth of resources and expert manpower.

CONCLUSION

Early detection and optimal management of LC are instrumental in strengthening the robust cancer care and control system in the country. In India, there is an unmet need to decrease the lag period from symptom onset to initiation of treatment of LC compared to Western countries. Achieving early diagnosis of LC requires GPs to maintain a high level of suspicion and readiness to investigate patients at high-risk or those with non-resolving symptoms. Consultation with an MDT at the emergence of a red flag itself could be the key to providing a timely and accurate diagnosis and treatment of LC. CXRs and CT scans are the routine diagnostic imaging procedures utilised for suspected LC in primary care. Tissue diagnosis is the currently recommended approach and the gold standard for confirming the diagnosis of LC. AI can assess CXR and CT scans for incidental nodule identification in diverse settings, including primary care, acting as a facilitator for further evaluation. Incidental nodule detection programme in hospitals with MDT can be looked at as a complementary strategy to detect lung cancer. Proactive screening, referral to specialists, and aggressive follow-up can have the maximum impact on patient outcomes. Vigilant PCPs, early detection through wider adoption of LC screening in a high-risk population and improving access to cancer care are vital for the advancement of LC care in India.

Authors’ contributions

All named authors meet the International Committee of Medical Journal Editors criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Financial support and sponsorship

AstraZeneca Pharma India Ltd.

Conflicts of interest

There are no conflicts of interest.

SUPPLEMENTARY DATA

Supplementary Table S1.

Population-based estimates of the frequencies of presenting symptoms among lung cancer patients[9,30,33,34,42]

Study Study period Setting Sample size Study population Percentage (%) of occurrence of symptoms
Chandra et al.,[9] 2002-2008 Tertiary care 165 No limit Coughing: 75.2
Shortness of breath: 66.9
Weight loss: 63.7
Chest pain: 63.1
Haemoptysis: 33.1
Hoarseness of voice: 29.3
Excessive weakness or fatigue: 26.8
Clubbing: 22.9
Dysphagia: 9.3
SVC: 8.0
Walter et al., [30] 2010-2012 Primary and secondary care data; self-reported symptoms before diagnosis 153 40+years Haemoptysis: 21.6
Cough or worsening cough: 56.2
Breathlessness or worsening breathlessness: 41.2
Chest-shoulder pain: 35.3
Hoarseness: 12.4
Decreased appetite: 22.2
Unexplained weight loss: 15
Fatigue or tiredness: 45.1
Feeling different “in yourself”: 34.6
Gupta et al. [33] Not specified Tertiary care 170 No limit Cough: 90.0
Loss of appetite and weight loss: 80.0
Expectoration: 79.4
Non-specific constitutional symptoms: 74.7
Chest pain or discomfort: 67.6
Shortness of breath: 54.7
Fever: 42.3
Lymphadenopathy: 30
Haemoptysis: 28.2
Hoarseness of voice: 24.7
Neurological signs: 14.7
SVC syndrome: 14.1
Bone pain: 11.7
Puffiness of face: 9.4
Asymptomatic: 3.5
Subcutaneous nodules: 3.5
HPOA: 2.9
Dysphagia: 1.7
Horner’s syndrome: 1.7
Gynaecomastia: 1.7
Deep vein thrombosis: 1.1
Dubey et al. [34] 2012-2013 Tertiary care 62 No limit Cough: 80.5
Chest: 74.4
Dyspnoea: 61.7
Decreased appetite: 44.6
Haemoptysis: 36.1
Weight loss: 23.4
Hoarseness of voice: 10.6
Fever: 22.2
Swelling over the face: 10.6
Dysphagia: 8.5
Vomiting: 4.2
Body ache: 12.7
Weakness: 6.3
Hamilton et al.[42] 1998-2002 Primary care, data from 21 general practices 247 40+years Haemoptysis: 20
Weight loss: 27
Loss of appetite: 19
Dyspnoea: 56
Chest or rib pain: 42
Fatigue: 35
Finger clubbing: 4.5
Thrombocytosis: 14
Abnormal spirometry: 9.7

HPOA: Hypertrophic pulmonary osteoarthropathy, SVC: Superior vena cava

Supplementary Table S2.

Risk rate and positive predictive rate for lung cancer[42,43]

Symptoms Odds ratio 95% Confidence interval P value Positive predictive value
Loss of appetite 86 (3.6 to 2100), 0.006 0.87 (0.6, 1.3)
Haemoptysis 32 (13 to 81), <0.001 2.4 (1.4, 4.1)
Cough Not applicable 0.40 (0.3, 0.5)
Dyspnoea 4.7 (2.7 to 8.0), <0.001 0.66 (0.5, 0.8)
Loss of weight 4.3 (2.2 to 8.2), <0.001 1.1 (0.8, 1.6)
Fatigue 3.2 (1.7 to 6.0), <0.001 0.43 (0.3, 0.6)
Chest pain 2.9 (1.8 to 4.7), <0.001 0.82 (0.6, 1.1)
Finger clubbing 18 (1.7 to 190), 0.016 Not applicable
Thrombocytosis 9.3 (3.4 to 26), 0.001 1.6 (0.8, 3.1)
Abnormal spirometry 7.5 (2.8 to 21), 0.001 1.6 (0.9, 2.9)
Dyspnoea with fatigue 0.28 (0.11 to 0.73), 0.006 0.66 (0.5, 0.8)
Loss of appetite in patients over 70 years 0.13 (0.024 to 0.76), 0.02 0.87 (0.6, 1.3)

Supplementary Table S3.

Summary of clinical guidelines for evaluation and management of incidental solitary pulmonary nodules[54,57,58]

Name of the society Sub-solid nodules Small solid nodules Large solid nodules Comments
Fleischner Society[54] Solitary solid nodules <6 mm (<100 mm3)
 Low-risk patients: No routine follow-up required High-risk patients: Optional CT at 12 months (particularly with suspicious nodule morphology and/or upper lobe location) Sub-solid single GGN <6 mm (<100 mm3) No routine follow-up required Sub-solid single part solid nodule <6 mm (<100 mm3) No routine follow-up required
Solitary solid nodules 6-8 mm (100-250 mm3)
 Low-risk patients: CT at 6-12 months, then consider CT at 18-24 months High-risk patients: CT at 6-12 months, then CT at 18-24 months Sub-solid single GGN ≥6 mm (>100 mm3) CT at 6-12 months, then if persistent, CT every 2 years until 5 years Sub-solid single part solid nodule ≥6 mm (>100 mm3) CT at 3-6 months, then if persistent and solid component remains <6 mm, annual CT until 5 years
Solitary solid nodule >8 mm (>250 mm3)
 Low-risk and high-risk patients: Consider CT at 3 months, PET/CT, or tissue sampling
In low-risk patients: Nodules <6 mm do not require routine follow-up. Certain patients at high-risk with suspicious nodule morphology, upper lobe location, or both may warrant 12-month follow-up. In certain suspicious nodules, 6 mm, consider follow-up at 2 and 4 years. If solid component (s) or growth develops, consider resection. In practice, part solid nodules cannot be defined as such until ≥6 mm, and nodules, <6 mm do not usually require follow-up. Persistent part solid nodules with solid components ≥6 mm should be considered highly suspicious.
ACCP[57] Single GGN ≤5 mm
 No routine follow-up required Single GGN >5 mm
 Annual CT chest for 3 years Single GGN >10 mm
 Repeat CT in 3 months followed by a biopsy if persistent Single part solid nodule ≤8 mm
 CT surveillance in 3, 12, and 24 months followed by annual CT for an additional 1-3 years Single part solid nodule >8 mm CT chest in 3 months followed by
 PET/CT (if solid component >8 mm) or biopsy if persistent or growing Single part solid nodule >15 mm PET/CT or biopsy PET does not reliably identify malignant nodules in this group, particularly pure GGNs TTNB has suboptimal sensitivity High likelihood of malignancy and need for longer surveillance
Nodule <8 mm
 Interval follow-up CT based on size and individual risk factors for cancer (generally 6-12 months) Size divided into ≤4, 4-6, 6-8 Does not recommend functional imaging or biopsy given most nodules in this group are benign and unreliability of diagnostic modalities
Nodule ≥8 mm CT surveillance, PET/CT, nonsurgical biopsy, or surgical biopsy are appropriate depending on the probability of cancer, surgical risk, and patient preference In low-risk (<5%) patients: CT surveillance In intermediate risk (5%-65%) patients: PET/CT for further risk stratification In high-risk (>65%) patients: Biopsy or resection. No PET, unless staging TTNB or bronchoscopy modalities depending on nodule location and centre expertise Same algorithm for screening and incidental detected nodules. Low-risk <5%, intermediate risk 5%-65%, high-risk >65%. Low-risk defined as young, less smoking, no prior cancer, small nodule, regular margins and not located in upper lobes. High-risk is the opposite TTNB and intermediate features of both. Predictive model or clinical judgment appropriate (mentions Mayo as most validated). Solid nodule stable for ≥2 years; no follow-up.
BTS[58] Nodule <5 mm (80 μl)
 No follow-up required Nodule ≥5 mm CT at 3 months, and if stable, use Brock model. If risk <10%, then CT surveillance at 1, 2, and 4 years. If risk>10% consider biopsy, resection or CT surveillance. If growth at 3 months consider resection, nonsurgical treatment, or CT surveillance.
Nodule <5 mm (80 μl)
 No follow-up required Nodule 5-6 mm CT surveillance at 1 year. Further evaluation will depend on the stability 6-8 mm (80-300 μl) CT surveillance at 3 months. If VDT <400 or clear growth requires further investigation. If stable, repeat CT in 1 year
Nodules ≥8 mm (≥300 μl)
 Brock model for risk assessment If <10%, CT surveillance If >10%, PET/CT and apply Herder model If <10% after Herder, CT surveillance If 10%-70% after Herder, CT surveillance, or biopsybased on risk and individual preference If >70% after Herder, surgery, nonsurgical biopsy, or nonsurgical treatment
Same algorithm for screening and incidentally detected nodules Applies to all adults ≥18 years old, and nodules detected in the context of current or prior malignancy Recommends using Brock model and then PET/CT and Herder model if risk is >10%

ACCP: American College of Chest Physicians; BTS: British Thoracic Society; CT: Computed tomography; GGN: Ground glass nodule; IPN: Indeterminate pulmonary nodule; PET: Positron-emission tomography; TTNB: Transthoracic needle biopsy; VDT: Volume doubling time. Summary of clinical guidelines for single pulmonary nodule. In general, guidelines recommend evaluation based on dominant/larger nodule. Refer to individual guidelines for management of multiple nodules. These recommendations are for IPN [no features of benign (fat), and not calcified in benign pattern]. All guidelines recommend reviewing prior imaging when IPN first identified nodule

Acknowledgments

The authors would like to thank AstraZeneca Pharma India Ltd., for the development of this manuscript in collaboration with Dr. Sasikala Somara, Labcorp Scientific Services & Solutions Pvt. Ltd., in accordance with GPP3 guidelines (http://www.ismpp.org/gpp3).

REFERENCES

  • 1.The Global Cancer Observatory 2020. Factsheets, Lung Cancer. [Last accessed on 2021 Sep 13]. Available from: http://gco.iarc.fr/today/home .
  • 2.Mohan A, Garg A, Gupta A, Sahu S, Choudhari C, Vashistha V, et al. Clinical profile of lung cancer in North India: A 10-year analysis of 1862 patients from a tertiary care center. Lung India. 2020;37:190–7. doi: 10.4103/lungindia.lungindia_333_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Institute for Health Metrics and Evaluation. Global burden of disease 2019. [Last accessed on 2021 Sep 27]. Available from: http://vizhub.healthdata.org/gbd-compare .
  • 4.Noronha V, Dikshit R, Raut N, Joshi A, Pramesh CS, George K, et al. Epidemiology of lung cancer in India: Focus on the differences between non-smokers and smokers: A single-centre experience. Indian J Cancer. 2012;49:74–81. doi: 10.4103/0019-509X.98925. [DOI] [PubMed] [Google Scholar]
  • 5.Kaur H, Sehgal IS, Bal A, Gupta N, Behera D, Das A, et al. Evolving epidemiology of lung cancer in India: Reducing non-small cell lung cancer-not otherwise specified and quantifying tobacco smoke exposure are the key. Indian J Cancer. 2017;54:285–90. doi: 10.4103/ijc.IJC_597_16. [DOI] [PubMed] [Google Scholar]
  • 6.Allemani C, Weir HK, Carreira H, Harewood R, Spika D, Wang X-S, et al. Global surveillance of cancer survival 1995-2009: Analysis of individual data for 25 676 887 patients from 279 population-based registries in 67 countries (CONCORD-2) Lancet. 2015;385:977–1010. doi: 10.1016/S0140-6736(14)62038-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Shridhar K, Kapoor R, Goodman M, Kondal D, Narang K, Singh P, et al. Lung and gallbladder cancer survival in north India: An ambidirectional feasibility cohort study using telephone interviews. J Global Health Rep. 2020;4:e2020053. doi:10.29392/001c. 13074. [Google Scholar]
  • 8.Ramachandran K, Thankagunam B, Karuppusami R, Christopher D. Physician related delays in the diagnosis of lung cancer in India. J Clin Diagn Res. 2016;10:OC05–8. doi: 10.7860/JCDR/2016/22737.8823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chandra S, Mohan A, Guleria R, Singh V, Yadav P. Delays during the diagnostic evaluation and treatment of lung cancer. Asian Pac J Cancer Prev. 2009;10:453–6. [PubMed] [Google Scholar]
  • 10.National Lung Screening Trial Research Team. Aberle DR, Adams AM, Berg CD, Black WC, Clapp JD, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365:395–409. doi: 10.1056/NEJMoa1102873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WEE, et al. The IASLC lung cancer staging project: Proposals for revision of the TNM stage groupings in the forthcoming (Eighth) edition of the TNM classification for lung cancer. J Thorac Oncol. 2016;11:39–51. doi: 10.1016/j.jtho.2015.09.009. [DOI] [PubMed] [Google Scholar]
  • 12.Murali AN, Radhakrishnan V, Ganesan TS, Rajendranath R, Ganesan P, Selvaluxmy G, et al. Outcomes in lung cancer:9-year experience from a tertiary cancer center in India. J Glob Oncol. 2017;3:459–68. doi: 10.1200/JGO.2016.006676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kumar D, Singh S. Factors causing treatment delays and its impact on treatment outcome in patients of lung cancer: An analysis. Ann Oncol. 2017;28:ii7. [Google Scholar]
  • 14.Howington JA, Blum MG, Chang AC, Balekian AA, Murthy SC. Treatment of stage I and II non-small cell lung cancer. Chest. 2013;143:e278S–313S. doi: 10.1378/chest.12-2359. [DOI] [PubMed] [Google Scholar]
  • 15.Howlader N, Forjaz G, Mooradian MJ, Meza R, Kong CY, Cronin KA, et al. The effect of advances in lung-cancer treatment on population mortality. N Engl J Med. 2020;383:640–9. doi: 10.1056/NEJMoa1916623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Whitson BA, Groth SS, Duval SJ, Swanson SJ, Maddaus MA. Surgery for early-stage non-small cell lung cancer: A systematic review of the video-assisted thoracoscopic surgery versus thoracotomy approaches to lobectomy. Ann Thorac Surg. 2008;86:2008–16. doi: 10.1016/j.athoracsur.2008.07.009. discussion 2016-2018. [DOI] [PubMed] [Google Scholar]
  • 17.Wang L, Liu D, Lu J, Zhang S, Yang X. The feasibility and advantage of uniportal video-assisted thoracoscopic surgery (VATS) in pulmonary lobectomy. BMC Cancer. 2017;17:75. doi: 10.1186/s12885-017-3069-z. doi:10.1186/s12885-017-3069-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Jiang L, Liu J, Gonzalez-Rivas D, Shargall Y, Kolb M, Shao W, et al. Thoracoscopic surgery for tracheal and carinal resection and reconstruction under spontaneous ventilation. J Thorac Cardiovasc Surg. 2018;155:2746–54. doi: 10.1016/j.jtcvs.2017.12.153. [DOI] [PubMed] [Google Scholar]
  • 19.Hung W-T, Cheng Y-J, Chen J-S. Nonintubated thoracoscopic surgery for early-stage non-small cell lung cancer. Gen Thorac Cardiovasc Surg. 2020;68:733–9. doi: 10.1007/s11748-019-01220-5. [DOI] [PubMed] [Google Scholar]
  • 20.Kneuertz PJ, D’Souza DM, Richardson M, Abdel-Rasoul M, Moffatt-Bruce SD, Merritt RE. Long-term oncologic outcomes after robotic lobectomy for early-stage non-small-cell lung cancer versus video-assisted thoracoscopic and open thoracotomy approach. Clin Lung Cancer. 2020;21:214–24.e2. doi: 10.1016/j.cllc.2019.10.004. [DOI] [PubMed] [Google Scholar]
  • 21.Wisnivesky JP, Smith CB, Packer S, Strauss GM, Lurslurchachai L, Federman A, et al. Survival and risk of adverse events in older patients receiving postoperative adjuvant chemotherapy for resected stages II-IIIA lung cancer: Observational cohort study. BMJ. 2011;343:d4013. doi: 10.1136/bmj.d4013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lindberg K, Nyman J, Riesenfeld Källskog V, Hoyer M, Lund JÅ, Lax I, et al. Long-term results of a prospective phase II trial of medically inoperable stage I NSCLC treated with SBRT - the Nordic experience. Acta Oncol. 2015;54:1096–104. doi: 10.3109/0284186X.2015.1020966. [DOI] [PubMed] [Google Scholar]
  • 23.van den Berg LL, Klinkenberg TJ, Groen HJM, Widder J. Patterns of recurrence and survival after surgery or stereotactic radiotherapy for early stage NSCLC. J Thorac Oncol. 2015;10:826–31. doi: 10.1097/JTO.0000000000000483. [DOI] [PubMed] [Google Scholar]
  • 24.Dahele M, Senan S. The role of stereotactic ablative radiotherapy for early-stage and oligometastatic non-small cell lung cancer: Evidence for changing paradigms. Cancer Res Treat. 2011;43:75–82. doi: 10.4143/crt.2011.43.2.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Li Y, Appius A, Pattipaka T, Feyereislova A, Cassidy A, Ganti AK. Real-world management of patients with epidermal growth factor receptor (EGFR) mutation-positive non-small-cell lung cancer in the USA. PLoS One. 2019;14:e0209709. doi: 10.1371/journal.pone.0209709. doi:10.1371/journal.pone. 0209709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Shu CA, Gainor JF, Awad MM, Chiuzan C, Grigg CM, Pabani A, et al. Neoadjuvant atezolizumab and chemotherapy in patients with resectable non-small-cell lung cancer: An open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020;21:786–95. doi: 10.1016/S1470-2045(20)30140-6. [DOI] [PubMed] [Google Scholar]
  • 27.Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med. 2020;382:41–50. doi: 10.1056/NEJMoa1913662. [DOI] [PubMed] [Google Scholar]
  • 28.Birring S, Peake M. Symptoms and the early diagnosis of lung cancer. Thorax. 2005;60:268–9. doi: 10.1136/thx.2004.032698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Barros JA, Valladares G, Faria AR, Fugita EM, Ruiz AP, Vianna AGD, et al. Early diagnosis of lung cancer: The great challenge. Epidemiological variables, clinical variables, staging and treatment. J Bras Pneumol. 2006;32:221–7. [PubMed] [Google Scholar]
  • 30.Walter FM, Rubin G, Bankhead C, Morris HC, Hall N, Mills K, et al. Symptoms and other factors associated with time to diagnosis and stage of lung cancer: A prospective cohort study. Br J Cancer. 2015;112(Suppl 1):S6–13. doi: 10.1038/bjc.2015.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Shanthilal M, Sathya M. Factors contributing to delays in the management of lung cancer: Retrospective study from government cancer center in India. J Glob Oncol. 2018;4:93s. [Google Scholar]
  • 32.Singh VK, Chandra S, Kumar S, Pangtey G, Mohan A, Guleria R. A common medical error: Lung cancer misdiagnosed as sputum negative tuberculosis. Asian Pac J Cancer Prev. 2009;10:335–8. [PubMed] [Google Scholar]
  • 33.Gupta R, Chowdhary I, Singh P. Clinical, radiological and histological profile of primary lung carcinomas. JK Sci. 2015;17:6. [Google Scholar]
  • 34.Dubey N, Arti J, Varudkar HG, Agrawat JC, Bhandari D, Mukati S, et al. A clinico-pathological profile of primary lung cancer patients presenting in a rural medical college of Central India. Panacea J Med Sci. 2015;5:124–9. [Google Scholar]
  • 35.Vashistha V, Choudhari C, Garg A, Gupta A, Parthasarathy G, Jain D, et al. The time required to diagnose and treat lung cancer in Delhi, India: An updated experience of a public referral center. Appl Cancer Res. 2019;39:11. [Google Scholar]
  • 36.Wang H, Yang L, Zou L, Huang D, Guo Y, Pan M, et al. Association between chronic obstructive pulmonary disease and lung cancer: A case-control study in Southern Chinese and a meta-analysis. PLoS One. 2012;7:e46144. doi: 10.1371/journal.pone.0046144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Brenner DR, McLaughlin JR, Hung RJ. Previous lung diseases and lung cancer risk: A systematic review and meta-analysis. PLoS One. 2011;6:e17479. doi: 10.1371/journal.pone.0017479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wait S & The health policy partnership. Lung Cancer Screening: Building Resilience and Sustainability of Healthcare Systems. London: HPP; 2021. [Google Scholar]
  • 39.Chaturvedi P, Chaturvedi U. Changes needed for improved cancer care in the developing world. Lancet Oncol. 2002;3:526–7. doi: 10.1016/s1470-2045(02)00843-4. [DOI] [PubMed] [Google Scholar]
  • 40.Triphuridet N, Henschke C. Landscape on CT screening for lung cancer in Asia. Lung Cancer (Auckl) 2019;10:107–24. doi: 10.2147/LCTT.S192643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cassidy A, Myles JP, van Tongeren M, Page RD, Liloglou T, Duffy SW, et al. The LLP risk model: An individual risk prediction model for lung cancer. Br J Cancer. 2008;98:270–6. doi: 10.1038/sj.bjc.6604158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hamilton W, Peters TJ, Round A, Sharp D. What are the clinical features of lung cancer before the diagnosis is made?A population based case-control study. Thorax. 2005;60:1059–65. doi: 10.1136/thx.2005.045880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bradley SH, Kennedy MPT, Neal RD. Recognising lung cancer in primary care. Adv Ther. 2019;36:19–30. doi: 10.1007/s12325-018-0843-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cancer Australia, 2020. Evidence report for Investigating symptoms of lung cancer: A guide for all health professionals, Cancer Australia, Surry Hills, NSW. [Last accessed on 2022 May 19]. Available from: https://www.canceraustralia.gov.au/sites/default/files/publications/evidence-report-investigating-symptomslung-cancer-guide-all-health-professionals/pdf/investigating_symptoms_of_lung_cancer_evidence_report_june_2020.pdf .
  • 45.Bilfinger TV, Albano D, Perwaiz M, Keresztes R, Nemesure B. Survival outcomes among lung cancer patients treated using a multidisciplinary team approach. Clin Lung Cancer. 2018;19:346–51. doi: 10.1016/j.cllc.2018.01.006. [DOI] [PubMed] [Google Scholar]
  • 46.Pan C-C, Kung P-T, Wang Y-H, Chang Y-C, Wang S-T, Tsai W-C. Effects of multidisciplinary team care on the survival of patients with different stages of non-small cell lung cancer: A national cohort study. PLoS One. 2015;10:e0126547. doi: 10.1371/journal.pone.0126547. doi:10.1371/journal.pone. 0126547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Stone E, Rankin N, Currow D, Fong KM, Phillips JL, Shaw T. Optimizing lung cancer MDT data for maximum clinical impact—A scoping literature review. Transl Lung Cancer Res. 2020;9:1629–38. doi: 10.21037/tlcr.2020.01.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Griffith C, Turner J. United Kingdom National Health Service. Cancer services collaborative “improvement partnership”,: Redesign of cancer services A national approach. Eur J Surg Oncol. 2004;30:1–86. doi: 10.1016/j.ejso.2004.07.010. [DOI] [PubMed] [Google Scholar]
  • 49.Denton E, Conron M. Improving outcomes in lung cancer:The value of the multidisciplinary health care team. J Multidiscip Healthc. 2016;9:137–44. doi: 10.2147/JMDH.S76762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Deepak JA, Ng X, Feliciano J, Mao L, Davidoff AJ. Pulmonologist involvement, stage-specific treatment, and survival in adults with non-small cell lung cancer and chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2015;12:742–51. doi: 10.1513/AnnalsATS.201406-230OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hussain T, Chang H-Y, Veenstra CM, Pollack CE. Collaboration between surgeons and medical oncologists and outcomes for patients with stage III colon cancer. J Oncol Pract. 2015;11:e388–97. doi: 10.1200/JOP.2014.003293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rogers SO, Ayanian JZ, Ko CY, Kahn KL, Zaslavsky AM, Sandler RS, et al. Surgeons'volume of colorectal cancer procedures and collaborative decision-making about adjuvant therapies. Ann Surg. 2009;250:895–900. doi: 10.1097/SLA.0b013e3181afe0c6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Prabhakar CN, Fong KM, Peake MD, Lam DC, Barnes DJ. The effectiveness of lung cancer MDT and the role of respiratory physicians. Respirology. 2015;20:884–8. doi: 10.1111/resp.12520. [DOI] [PubMed] [Google Scholar]
  • 54.MacMahon H, Naidich DP, Goo JM, Lee KS, Leung ANC, Mayo JR, et al. Guidelines for Management of incidental pulmonary nodules detected on CT images: From the Fleischner Society 2017. Radiology. 2017;284:228–43. doi: 10.1148/radiol.2017161659. [DOI] [PubMed] [Google Scholar]
  • 55.Asija A, Manickam R, Aronow WS, Chandy D. Pulmonary nodule: A comprehensive review and update. Hosp Pract. 2014;42:7–16. doi: 10.3810/hp.2014.08.1125. [DOI] [PubMed] [Google Scholar]
  • 56.Wahidi MM, Govert JA, Goudar RK, Gould MK, McCrory DC American College of Chest Physicians. Evidence for the treatment of patients with pulmonary nodules: When is it lung cancer?: ACCP evidence-based clinical practice guidelines (2nd edition) Chest. 2007;132:94S–107. doi: 10.1378/chest.07-1352. [DOI] [PubMed] [Google Scholar]
  • 57.Gould MK, Donington J, Lynch WR, Mazzone PJ, Midthun DE, Naidich DP, et al. Evaluation of individuals with pulmonary nodules: When is it lung cancer? Chest. 2013;143:e93S–e120. doi: 10.1378/chest.12-2351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Baldwin DR, Callister MEJ Guideline Development Group. The British Thoracic Society guidelines on the investigation and management of pulmonary nodules. Thorax. 2015;70:794–8. doi: 10.1136/thoraxjnl-2015-207221. [DOI] [PubMed] [Google Scholar]
  • 59.Bradley SH, Abraham S, Callister ME, Grice A, Hamilton WT, Lopez RR, et al. Sensitivity of chest X-ray for detecting lung cancer in people presenting with symptoms: A systematic review. Br J Gen Pract. 2019;69:e827–35. doi: 10.3399/bjgp19X706853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bradley S, Bradley S, Abraham S, Grice A, Lopez RR, Wright J, et al. Sensitivity of chest X-ray for lung cancer: Systematic review. [Last accessed on 2021 Sep 16];Br J Gen Pract. 2018 68 Available from: https://bjgp.org/content/68/suppl_1/bjgp18x696905 . [Google Scholar]
  • 61.Quekel LG, Kessels AG, Goei R, van Engelshoven JM. Miss rate of lung cancer on the chest radiograph in clinical practice. Chest. 1999;115:720–4. doi: 10.1378/chest.115.3.720. [DOI] [PubMed] [Google Scholar]
  • 62.del Ciello A, Franchi P, Contegiacomo A, Cicchetti G, Bonomo L, Larici AR. Missed lung cancer: When, where, and why? Diagn Interv Radiol. 2017;23:118–26. doi: 10.5152/dir.2016.16187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Winer-Muram HT. The solitary pulmonary nodule. Radiology. 2006;239:34–49. doi: 10.1148/radiol.2391050343. [DOI] [PubMed] [Google Scholar]
  • 64.Swensen SJ, Jett JR, Hartman TE, Midthun DE, Sloan JA, Sykes A-M, et al. Lung cancer screening with CT: Mayo Clinic experience. Radiology. 2003;226:756–61. doi: 10.1148/radiol.2263020036. [DOI] [PubMed] [Google Scholar]
  • 65.Zhang Y-J, Yang W-J, Liu D, Cao Y-Q, Zheng Y-Y, Han Y-C, et al. COVID-19 and early-stage lung cancer both featuring ground-glass opacities: A propensity score-matched study. Transl Lung Cancer Res. 2020;9:1516–27. doi: 10.21037/tlcr-20-892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gao F, Li M, Sun Y, Xiao L, Hua Y. Diagnostic value of contrast-enhanced CT scans in identifying lung adenocarcinomas manifesting as GGNs (ground glass nodules) Medicine (Baltimore) 2017;96:e7742. doi: 10.1097/MD.0000000000007742. doi:10.1097/MD.0000000000007742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Garcia-Velloso MJ, Bastarrika G, de-Torres JP, Lozano MD, Sanchez-Salcedo P, Sancho L, et al. Assessment of indeterminate pulmonary nodules detected in lung cancer screening: Diagnostic accuracy of FDG PET/CT. Lung Cancer. 2016;97:81–6. doi: 10.1016/j.lungcan.2016.04.025. [DOI] [PubMed] [Google Scholar]
  • 68.Purandare NC, Pramesh CS, Agarwal JP, Agrawal A, Shah S, Prabhash K, et al. Solitary pulmonary nodule evaluation in regions endemic for infectious diseases:Do regional variations impact the effectiveness of fluorodeoxyglucose positron emission tomography/computed tomography. Indian J Cancer. 2017;54:271–5. doi: 10.4103/0019-509X.219563. [DOI] [PubMed] [Google Scholar]
  • 69.Kumar A, Dutta R, Kannan U, Kumar R, Khilnani GC, Gupta SD. Evaluation of mediastinal lymph nodes using F-FDG PET-CT scan and its histopathologic correlation. Ann Thorac Med. 2011;6:11–6. doi: 10.4103/1817-1737.74270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Madan K, Mohan A, Agarwal R, Hadda V, Khilnani GC, Guleria R. A survey of flexible bronchoscopy practices in India: The Indian bronchoscopy survey (2017) Lung India. 2018;35:98–107. doi: 10.4103/lungindia.lungindia_417_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Madhusudhan KS, Srivastava DN, Sharma S, Sharma S. Interventional radiology in India. Am J Roentgenol. 2018;211:730–5. doi: 10.2214/AJR.18.19777. [DOI] [PubMed] [Google Scholar]
  • 72.Radhakrishnan RK, Mittal BR, Gorla AKR, Basher RK, Sood A, Bal A, et al. Real-time intraprocedural 18F-FDG PET/CT-guided biopsy using automated robopsy arm (ARA) in the diagnostic evaluation of thoracic lesions with prior inconclusive biopsy results: Initial experience from a tertiary health care centre. Br J Radiol. 2017;90:20170258. doi: 10.1259/bjr.20170258. doi:10.1259/bjr. 20170258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Apurva A, Tandon SP, Shetmahajan M, Jiwnani SS, Karimundackal G, Pramesh CS. Surgery for lung cancer—the Indian scenario. Indian J Thorac Cardiovasc Surg. 2018;34:47–53. [Google Scholar]
  • 74.Sehgal IS, Dhooria S, Aggarwal AN, Behera D, Agarwal R. Endosonography versus mediastinoscopy in mediastinal staging of lung cancer: Systematic review and meta-analysis. Ann Thorac Surg. 2016;102:1747–55. doi: 10.1016/j.athoracsur.2016.05.110. [DOI] [PubMed] [Google Scholar]
  • 75.Wozniak MB, Scelo G, Muller DC, Mukeria A, Zaridze D, Brennan P. Circulating MicroRNAs as non-invasive biomarkers for early detection of non-small-cell lung cancer. PLoS One. 2015;10:e0125026. doi: 10.1371/journal.pone.0125026. doi:10.1371/journal.pone. 0125026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Liang W, Zhao Y, Huang W, Gao Y, Xu W, Tao J, et al. Non-invasive diagnosis of early-stage lung cancer using high-throughput targeted DNA methylation sequencing of circulating tumor DNA (ctDNA) Theranostics. 2019;9:2056–70. doi: 10.7150/thno.28119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Duan G-C, Zhang X-P, Wang H-E, Wang Z-K, Zhang H, Yu L, et al. Circulating tumor cells as a screening and diagnostic marker for early-stage non-small cell lung cancer. Onco Targets Ther. 2020;13:1931–9. doi: 10.2147/OTT.S241956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Mehta A, Anuj P, Bhatia M. Cross-sectional study to establish the utility of serum tumor markers in the diagnosis of lung cancer. Asian Pac J Cancer Prev. 2021;22:2569–76. doi: 10.31557/APJCP.2021.22.8.2569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kris MG, Johnson BE, Berry LD, Kwiatkowski DJ, Iafrate AJ, Wistuba II, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA. 2014;311:1998. doi: 10.1001/jama.2014.3741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kulkarni SS, Rauthan A, Patil P, Sood T, Somashekhar S, Zaveri S. Oncogenic driver mutation analysis in lung adenocarcinoma: A single center study in India. Ann Oncol. 2018;29:i–156. doi:10.1093/annonc/mdy425.019. [Google Scholar]
  • 81.Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Non-Small Cell Lung Cancer V.1.2022.© National Comprehensive Cancer Network, Inc. 2021. All rights reserved. [Last accessed on 2022 Jan 05]. To view the most recent and complete version of the guideline, go online to NCCN.org. NCCN makes no warranties of any kind whatsoever regarding their content, use or application and disclaims any responsibility for their application or use in any way.
  • 82.Kalemkerian GP, Narula N, Kennedy EB, Biermann WA, Donington J, Leighl NB, et al. Molecular testing guideline for the selection of patients with lung cancer for treatment with targeted tyrosine kinase inhibitors: American Society of Clinical Oncology Endorsement of the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology Clinical Practice Guideline Update. J Clin Oncol. 2018;36:911–9. doi: 10.1200/JCO.2017.76.7293. [DOI] [PubMed] [Google Scholar]
  • 83.Prabhash K, Advani S, Batra U, Biswas B, Chougule A, Ghosh M, et al. Biomarkers in non-small cell lung cancers: Indian consensus guidelines for molecular testing. Adv Ther. 2019;36:766–85. doi: 10.1007/s12325-019-00903-y. [DOI] [PubMed] [Google Scholar]
  • 84.Nakra T, Mehta A, Bal A, Nambirajan A, Mishra D, Midha D, et al. Epidermal growth factor receptor mutation status in pulmonary adenocarcinoma: Multi-institutional data discussion at national conference of “Lung Cancer Management in Indian context”. Curr Problm Cancer. 2020;44:100561. doi: 10.1016/j.currproblcancer.2020.100561. doi:10.1016/j.currproblcancer. 2020.100561. [DOI] [PubMed] [Google Scholar]
  • 85.Singh N, Agrawal S, Jiwnani S, Khosla D, Malik PS, Mohan A, et al. Lung cancer in India. J Thorac Oncol. 2021;16:1250–66. doi: 10.1016/j.jtho.2021.02.004. [DOI] [PubMed] [Google Scholar]
  • 86.Singh N. Lung cancer in India: The rewards of REWERDS (REal World Evidence from Retrospective Data analysiS) Curr Probl Cancer. 2020;44:100591. doi: 10.1016/j.currproblcancer.2020.100591. doi:10.1016/j.currproblcancer. 2020.100591. [DOI] [PubMed] [Google Scholar]
  • 87.Kumar M, Guleria B, Swamy S, Soni S. Correlation of programmed death-ligand 1 expression with gene expression and clinicopathological parameters in Indian patients with non-small cell lung cancer. Lung India. 2020;37:145–50. doi: 10.4103/lungindia.lungindia_488_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.US Preventive Services Task Force. Krist AH, Davidson KW, Mangione CM, Barry MJ, Cabana M, et al. Screening for lung cancer: US Preventive Services Task Force Recommendation Statement. JAMA. 2021;325:962–70. doi: 10.1001/jama.2021.1117. [DOI] [PubMed] [Google Scholar]
  • 89.Horeweg N, Aalst CM van der, Vliegenthart R, Zhao Y, Xie X, Scholten ET, et al. Volumetric computed tomography screening for lung cancer: Three rounds of the NELSON trial. Eur Respir J. 2013;42:1659–67. doi: 10.1183/09031936.00197712. [DOI] [PubMed] [Google Scholar]
  • 90.Handy JR, Skokan M, Rauch E, Zinck S, Sanborn RE, Kotova S, et al. Results of lung cancer screening in the community. Ann Fam Med. 2020;18:243–9. doi: 10.1370/afm.2519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Field JK, Duffy SW, Baldwin DR, Whynes DK, Devaraj A, Brain KE, et al. UK lung cancer RCT pilot screening trial: Baseline findings from the screening arm provide evidence for the potential implementation of lung cancer screening. Thorax. 2016;71:161–70. doi: 10.1136/thoraxjnl-2015-207140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.International Early Lung Cancer Action Program Investigators. Henschke CI, Yankelevitz DF, Libby DM, Pasmantier MW, Smith JP, et al. Survival of patients with stage I lung cancer detected on CT screening. N Engl J Med. 2006;355:1763–71. doi: 10.1056/NEJMoa060476. [DOI] [PubMed] [Google Scholar]
  • 93.Bagcchi S. India launches plan for national cancer screening programme. BMJ. 2016;355:i5574. doi: 10.1136/bmj.i5574. doi:10.1136/bmj.i5574. [DOI] [PubMed] [Google Scholar]
  • 94.Nam JG, Park S, Hwang EJ, Lee JH, Jin K-N, Lim KY, et al. Development and validation of deep learning-based automatic detection algorithm for malignant pulmonary nodules on chest radiographs. Radiology. 2019;290:218–28. doi: 10.1148/radiol.2018180237. [DOI] [PubMed] [Google Scholar]
  • 95.Zhang C, Sun X, Dang K, Li K, Guo X-W, Chang J, et al. Toward an expert level of lung cancer detection and classification using a deep convolutional neural network. Oncologist. 2019;24:1159–65. doi: 10.1634/theoncologist.2018-0908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lee JH, Sun HY, Park S, Kim H, Hwang EJ, Goo JM, et al. Performance of a deep learning algorithm compared with radiologic interpretation for lung cancer detection on chest radiographs in a health screening population. Radiology. 2020;297:687–96. doi: 10.1148/radiol.2020201240. [DOI] [PubMed] [Google Scholar]
  • 97.Liu X, Zhou H, Hu Z, Jin Q, Wang J, Ye B. Clinical application of artificial intelligence recognition technology in the diagnosis of stage T1 lung cancer. Zhongguo Fei Ai Za Zhi. 2019;22:319–23. doi: 10.3779/j.issn.1009-3419.2019.05.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Cui S, Ming S, Lin Y, Chen F, Shen Q, Li H, et al. Development and clinical application of deep learning model for lung nodules screening on CT images. Sci Rep. 2020;10:13657. doi: 10.1038/s41598-020-70629-3. doi:10.1038/s41598-020-70629-3. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Lung India : Official Organ of Indian Chest Society are provided here courtesy of Wolters Kluwer -- Medknow Publications

RESOURCES