1. Introduction
Lung cancer, chronic obstructive pulmonary disease (COPD), cardiovascular disease (CVD), and pulmonary fibrosis are among the most significant contributors to global morbidity and mortality. These diseases, which individually present substantial public health challenges, are frequently found to coexist in patients, creating a complex web of clinical interdependencies. While traditionally studied as separate entities, emerging evidence underscores the existence of shared risk factors, overlapping molecular mechanisms, and reciprocal influences that exacerbate disease progression and complicate treatment (Barnes et al., 2017; Hernandez et al., 2020).
Smoking remains the most prominent and well-established shared risk factor across these conditions. Tobacco smoke contains over 7,000 harmful chemicals that induce oxidative stress, chronic inflammation, and DNA damage, all of which are known to contribute to the development of lung cancer, COPD, CVD, and pulmonary fibrosis (Lee et al., 2017; Wouters et al., 2019). However, beyond smoking, other factors such as environmental pollution, genetic susceptibility, occupational exposure to hazardous substances, and aging play significant roles in disease onset and progression (Miller et al., 2016; Spira et al., 2020). For instance, exposure to particulate matter (PM2.5) and other airborne pollutants has been linked to chronic lung injury, cardiovascular dysfunction, and fibrotic lung disease. Additionally, genetic polymorphisms in key genes such as TERT, MUC5B, and TP53 have been implicated in the susceptibility to multiple pulmonary diseases, further highlighting the biological connections among these conditions (Willis et al., 2005; Spira et al., 2020).
Aging represents a crucial determinant in the development of these diseases, given that the aging process is accompanied by immune senescence, telomere attrition, and a decline in the body’s ability to repair damaged tissues. Age-related changes in immune responses and tissue repair mechanisms are critical drivers of chronic inflammation, a hallmark feature of lung cancer, COPD, CVD, and pulmonary fibrosis (Barnes et al., 2017; Chung et al., 2018). The concept of inflammaging, characterized by the chronic, low-grade inflammation that occurs with age, is increasingly recognized as a unifying factor contributing to multimorbidity in older adults. As such, the elderly population is disproportionately affected by these diseases, often facing poorer prognoses and greater healthcare burdens.
The interplay between lung cancer, COPD, CVD, and pulmonary fibrosis is not merely a result of shared risk factors but also reflects deeper biological and molecular interconnections. Chronic inflammation, oxidative stress, hypoxia, and epithelial-mesenchymal transition (EMT) are critical molecular mechanisms that operate across all four diseases (Hernandez et al., 2020; Barnes et al., 2017). For example, oxidative stress not only drives lung tissue damage in COPD and pulmonary fibrosis but also promotes genetic mutations and cancer progression. Similarly, EMT, which transforms epithelial cells into mesenchymal-like cells, is a key feature of tissue fibrosis and cancer metastasis (Willis et al., 2005). Transforming growth factor-beta (TGF-β) signaling, a pathway central to EMT, plays a pivotal role in fibrosis, tumor progression, and cardiovascular remodeling, making it a prime target for therapeutic intervention (Willis et al., 2005; Chung et al., 2018).
The coexistence of these diseases poses significant diagnostic and therapeutic challenges. Patients often present with overlapping symptoms such as dyspnea, chronic cough, and chest pain, complicating differential diagnosis. Imaging studies, including chest CT and PET-CT, can aid in differentiating these conditions. Misdiagnosis or delayed detection may lead to suboptimal treatment outcomes. Furthermore, the coexistence of multiple diseases within the same patient necessitates a more holistic approach to management. Pharmacological treatments, which are typically designed to address one condition at a time, may be inadequate or contraindicated in patients with multimorbidity, emphasizing the need for integrated treatment strategies (Wouters et al., 2019). For instance, corticosteroids commonly used in COPD management may worsen osteoporosis and cardiovascular risk, while antifibrotic drugs used in idiopathic pulmonary fibrosis (IPF) may have off-target effects on other organ systems.
Given the increasing burden of multimorbidity, it is essential to understand the intricate relationships among lung cancer, COPD, CVD, and pulmonary fibrosis. This review examines the epidemiological burden, shared pathophysiological mechanisms, and clinical implications of these coexisting diseases. We emphasize the importance of risk stratification, precision medicine, and the development of integrated therapeutic strategies aimed at improving patient outcomes. By addressing knowledge gaps and prioritizing future research, we aim to provide a comprehensive framework for understanding multimorbidity in lung diseases and its impact on patient care and healthcare systems worldwide
2. Epidemiology and Coexistence
2.1 Lung Cancer
Lung cancer is the leading cause of cancer-related deaths worldwide, with over 2.2 million new cases annually (Sung et al., 2021). It remains a significant contributor to the global cancer burden due to its high mortality rate and the complexity of treatment. Lung cancer frequently coexists with COPD, cardiovascular disease, and pulmonary fibrosis, creating diagnostic and therapeutic challenges. COPD is present in approximately 40-70% of lung cancer cases, underscoring a strong link between these conditions (Hernandez et al., 2020). The chronic inflammation and oxidative stress seen in COPD contribute to lung carcinogenesis through mechanisms such as increased production of reactive oxygen species (ROS) and activation of nuclear factor-kappa B (NF-κB), which promotes tumor growth and resistance to apoptosis (Barnes et al., 2017). Moreover, studies have highlighted that patients with idiopathic pulmonary fibrosis (IPF) have a 7-10% lifetime risk of developing lung cancer (Gillespie et al., 2022). The shared molecular pathways, such as epithelial-mesenchymal transition (EMT) and TGF-β signaling, underscore the biological interconnection between IPF and lung cancer (Willis et al., 2005).
2.2 Chronic Obstructive Pulmonary Disease (COPD)
COPD is a progressive lung disease that affects over 300 million people globally (GBD Chronic Respiratory Disease Collaborators, 2020). It is a significant risk factor for lung cancer due to its shared pathophysiological mechanisms, such as chronic inflammation, oxidative stress, and epithelial injury. Studies suggest that patients with COPD are four to six times more likely to develop lung cancer than non-COPD patients (Lee et al., 2017). Chronic inflammation in COPD, driven by elevated cytokines such as IL-6, IL-8, and TNF-α, increases the risk of cancerous transformation in lung epithelial cells. Furthermore, COPD is associated with systemic inflammation, which extends beyond the lungs to impact cardiovascular health. Endothelial dysfunction, characterized by reduced nitric oxide bioavailability and vascular stiffness, increases the risk of cardiovascular disease in COPD patients (Wouters et al., 2019). This shared risk factor necessitates a multidisciplinary approach to treatment, as therapeutic strategies for COPD may impact cardiovascular health and vice versa.
2.3 Cardiovascular Disease (CVD)
Cardiovascular disease (CVD) is a leading comorbidity in patients with COPD and lung cancer. It is responsible for a large proportion of morbidity and mortality in this population. Shared risk factors such as smoking, sedentary lifestyles, and systemic inflammation drive the development of CVD in COPD and lung cancer patients (Wouters et al., 2019). Chronic inflammation, characterized by elevated C-reactive protein (CRP) and pro-inflammatory cytokines, promotes endothelial dysfunction and accelerates atherosclerosis (Libby et al., 2002). This process is exacerbated by oxidative stress, which damages vascular endothelial cells and increases plaque formation in arteries. COPD patients have an increased risk of myocardial infarction, stroke, and heart failure, and this risk is further amplified in patients with lung cancer (Barnes et al., 2017). The use of certain cancer treatments, such as immune checkpoint inhibitors (ICIs) and chemotherapeutic agents, can induce cardiotoxic effects, further complicating the management of multimorbid patients.
2.4 Pulmonary Fibrosis
Pulmonary fibrosis, particularly idiopathic pulmonary fibrosis (IPF), frequently coexists with lung cancer and COPD. IPF is a chronic, progressive lung disease with an estimated prevalence of 13-20 per 100,000 individuals worldwide (Hutchinson et al., 2015). Patients with IPF are at significantly higher risk of developing lung cancer, with studies reporting a 7-10% lifetime risk of lung cancer in individuals diagnosed with IPF (Gillespie et al., 2022). The pathophysiology of IPF shares multiple similarities with lung cancer, particularly in terms of epithelial-mesenchymal transition (EMT) and TGF-β signaling (Willis et al., 2005). EMT promotes the transformation of epithelial cells into mesenchymal cells, a process that underlies both fibrotic remodeling in IPF and the metastatic potential of cancer cells. TGF-β, a key regulator of EMT, is upregulated in both IPF and lung cancer, making it a promising therapeutic target for both diseases. Moreover, the chronic inflammatory microenvironment in IPF facilitates genetic instability and enhances the likelihood of oncogenic mutations in lung epithelial cells (Willis et al., 2005). The coexistence of IPF and lung cancer necessitates a more nuanced clinical approach, as therapies like antifibrotic agents (e.g., pirfenidone and nintedanib) may have beneficial effects in mitigating cancer progression.
3. Shared Risk Factors
3.1 Smoking
Smoking is the most prominent shared risk factor for lung cancer, COPD, cardiovascular disease (CVD), and pulmonary fibrosis. Tobacco smoke contains over 7,000 toxic chemicals, many of which induce oxidative stress, DNA damage, and chronic inflammation (Barnes et al., 2017). The harmful components of cigarette smoke, such as nicotine, formaldehyde, benzene, and polycyclic aromatic hydrocarbons (PAHs), activate oxidative stress pathways by increasing reactive oxygen species (ROS) production, which damages cellular DNA, lipids, and proteins (Wouters et al., 2019). The chronic inflammation induced by cigarette smoke also results in the recruitment of immune cells, particularly neutrophils and macrophages, which release proteases and cytokines that further damage lung tissue. This cycle of injury, inflammation, and repair increases the likelihood of genetic mutations and epithelial-to-mesenchymal transition (EMT), facilitating the development of lung cancer and pulmonary fibrosis (Willis et al., 2005). Smoking also promotes atherosclerosis, endothelial dysfunction, and vascular inflammation, which are critical drivers of CVD (Libby et al., 2002). Importantly, smoking cessation has been shown to significantly reduce the risk of developing these diseases, particularly for lung cancer, where a 50% reduction in lung cancer risk is observed after five years of cessation (U.S. Surgeon General Report, 2020).
3.2 Environmental Pollutants
Environmental pollutants, including airborne particulate matter (PM2.5) and occupational exposure to chemicals and dust, increase the risk of developing lung cancer, COPD, pulmonary fibrosis, and cardiovascular disease. Prolonged exposure to these pollutants triggers oxidative stress, chronic inflammation, and lung injury, promoting the development and progression of chronic lung diseases (Miller et al., 2016). Particulate matter (PM2.5), a key component of air pollution, can penetrate deep into the alveoli of the lungs and enter the bloodstream, causing systemic inflammation and cardiovascular dysfunction (Miller et al., 2016). This exposure increases the risk of CVD, such as myocardial infarction and stroke, and exacerbates pre-existing respiratory diseases like COPD and IPF. Inhalation of occupational dust, including silica, asbestos, and coal dust, is also strongly linked to pulmonary fibrosis, leading to the development of pneumoconiosis and an increased risk of lung cancer (Hutchinson et al., 2015). Workplace exposure to organic dust and industrial fumes has been implicated in the pathogenesis of COPD, where inhalation of these irritants leads to chronic bronchitis and emphysema. Regulatory measures aimed at reducing air pollution and workplace exposure to hazardous materials have been effective in lowering the incidence of these diseases, particularly in industrial regions.
3.3 Genetic Susceptibility
Genetic susceptibility plays a significant role in predisposing individuals to lung cancer, COPD, pulmonary fibrosis, and CVD. Polymorphisms and mutations in genes such as MUC5B, TERT, TP53, and EGFR are associated with an increased risk of multiple conditions simultaneously (Spira et al., 2020). For example, the MUC5B promoter variant, which is a major genetic risk factor for idiopathic pulmonary fibrosis (IPF), has also been linked to an increased risk of lung cancer, particularly adenocarcinoma (Spira et al., 2020). The TERT gene, which encodes a key component of telomerase, is implicated in both telomere maintenance and cellular senescence. Mutations in TERT predispose individuals to pulmonary fibrosis, lung cancer, and other age-related diseases (Hutchinson et al., 2015). Similarly, mutations in the tumor suppressor gene TP53 are frequently observed in lung cancer and have been linked to poor prognosis due to its role in cell cycle regulation and apoptosis (Barnes et al., 2017). Polymorphisms in EGFR are particularly relevant for lung cancer, as they serve as both risk factors and therapeutic targets for targeted tyrosine kinase inhibitors (TKIs) (Willis et al., 2005). In addition to individual gene polymorphisms, whole-genome association studies (GWAS) have identified shared genetic loci that predispose individuals to multiple chronic diseases, further underscoring the genetic connections between COPD, lung cancer, and IPF. Genetic testing and risk stratification based on these susceptibility markers could pave the way for precision medicine and earlier interventions in high-risk populations.
3.4 Aging
Aging is a critical determinant of susceptibility to chronic diseases, including lung cancer, chronic obstructive pulmonary disease (COPD), cardiovascular disease (CVD), and pulmonary fibrosis. As individuals age, cellular and tissue homeostasis is disrupted by mechanisms such as immunosenescence, oxidative stress, and cellular senescence (Barnes et al., 2024; Childs et al., 2015).
Immunosenescence refers to the gradual decline of immune system function with age, characterized by a reduction in naive T cells and a shift toward a pro-inflammatory state, often referred to as “inflammaging” (Franceschi et al., 2018). This chronic, low-grade inflammation is a hallmark of aging and has been implicated in the pathogenesis of cancer, cardiovascular disease, and chronic lung disease (Libby et al., 2002). The pro-inflammatory environment driven by immunosenescence results in the sustained activation of immune cells, particularly macrophages, which produce cytokines such as IL-6 and TNF-\u03b1, thereby promoting tissue damage, fibrosis, and cancer progression (Rodier & Campisi, 2011; Franceschi et al., 2018).
Cellular senescence plays a crucial role in age-related tissue dysfunction. It is driven by mechanisms such as DNA damage, oxidative stress, and telomere shortening, which result in the secretion of pro-inflammatory cytokines, chemokines, and growth factors collectively known as the senescence-associated secretory phenotype (SASP) (van Deursen, 2014; Childs et al., 2015). The SASP drives fibrosis, angiogenesis, and tumor growth in the lungs, further linking aging to idiopathic pulmonary fibrosis (IPF), lung cancer, and CVD (Willis & Borok, 2005; Rodier & Campisi, 2011). Cellular senescence also promotes airway remodeling in COPD and vascular remodeling in CVD, further amplifying disease progression (Houssaini et al., 2018).
Telomere attrition, which occurs with aging, has been associated with an increased risk of lung cancer and IPF. As telomeres shorten with each cell division, their reduced length triggers DNA damage responses, leading to permanent cell cycle arrest (Chini et al., 2019). This senescence-like state not only affects lung epithelial cells but also impacts endothelial and immune cells, further promoting inflammation, fibrosis, and tumorigenesis (Kirkland & Tchkonia, 2017). Recent studies have linked telomere dysfunction to IPF and lung cancer, highlighting the need for therapeutic strategies targeting telomere maintenance (Hutchinson et al., 2015).
Oxidative stress further exacerbates age-related pathologies in the lungs and cardiovascular system. The age-related decline in antioxidant defense mechanisms, including reductions in superoxide dismutase (SOD) and glutathione (GSH) levels, enhances the production of reactive oxygen species (ROS) (Kinnula & Crapo, 2004; Kirkham & Barnes, 2013). Excessive ROS production damages mitochondrial DNA, lipids, and proteins, which in turn activates the DNA damage response and promotes cellular senescence. In COPD, oxidative stress is a key driver of emphysema, while in CVD, it promotes vascular endothelial dysfunction and atherosclerosis (Libby et al., 2002; Kirkham & Barnes, 2013).
As the global population ages, the burden of chronic lung diseases and multimorbidity is expected to rise. By 2050, the population aged 65 and older is projected to double, significantly increasing the healthcare burden (Barnes et al., 2024). This trend highlights the need for age-specific screening, preventive measures, and geriatric interventions aimed at reducing the impact of aging on chronic disease development. Research on anti-aging therapeutics, such as senolytics (drugs that selectively eliminate senescent cells), is underway, with the goal of mitigating the adverse effects of cellular senescence in chronic diseases (Kirkland & Tchkonia, 2017). Interventions targeting SASP components, such as IL-6 and TNF-\u03b1, are also being explored as potential therapies to reduce inflammation and fibrosis in age-related diseases (Childs et al., 2015; Franceschi et al., 2018).
4. Shared Pathophysiological Mechanisms
4.1 Inflammation
Chronic inflammation is a hallmark of lung cancer, COPD, cardiovascular disease (CVD), and pulmonary fibrosis. Persistent inflammation results from the continuous release of pro-inflammatory cytokines, chemokines, and reactive oxygen species (ROS) that drive pathological tissue remodeling, fibrosis, and carcinogenesis (Barnes et al., 2017). Key cytokines involved include IL-6, IL-8, and TNF-\u03b1, which promote tumor progression, airway remodeling, and endothelial dysfunction (Libby et al., 2002). IL-6, in particular, activates the STAT3 signaling pathway, enhancing cancer cell survival, proliferation, and immune evasion (Kang et al., 2019). In COPD, chronic exposure to cigarette smoke leads to recruitment of macrophages and neutrophils to the airways, which secrete proteases like matrix metalloproteinases (MMPs) and elastases, causing tissue destruction (Barnes et al., 2017).
In pulmonary fibrosis, macrophage polarization plays a key role in disease progression. M2 macrophages secrete pro-fibrotic cytokines such as TGF-\u03b2 and IL-13, promoting fibroblast activation and extracellular matrix (ECM) deposition (Willis et al., 2005). In CVD, low-grade chronic inflammation triggers endothelial dysfunction, a key driver of atherosclerosis. Elevated levels of C-reactive protein (CRP) and IL-6 correlate with plaque instability and cardiovascular events (Libby et al., 2002). Given the central role of inflammation in these diseases, targeting key mediators such as IL-6, TNF-\u03b1, and MMPs has emerged as a promising therapeutic approach to reduce disease burden.
4.2 Oxidative Stress
Oxidative stress, a state of imbalance between reactive oxygen species (ROS) production and antioxidant defense systems, is a central driver of disease progression in lung cancer, chronic obstructive pulmonary disease (COPD), cardiovascular disease (CVD), and pulmonary fibrosis. ROS are highly reactive molecules that damage cellular components such as DNA, proteins, and lipids, leading to genomic instability, tissue injury, and chronic inflammation (Barnes et al., 2022; Kinnula & Crapo, 2004).
In lung cancer, oxidative stress promotes the mutation of key oncogenes and tumor suppressor genes, including TP53, which is frequently altered in lung cancer (Barnes et al., 2022; Willis & Borok, 2007). ROS-induced DNA damage activates the DNA damage response (DDR) pathway, resulting in the accumulation of genetic alterations that drive carcinogenesis (Libby et al., 2002). The pro-oxidative environment also supports tumor survival and progression by activating pathways like NF-κB and HIF-1α, both of which are implicated in cancer proliferation, angiogenesis, and immune evasion (Rodier & Campisi, 2011; Oczypok et al., 2017).
In COPD, oxidative stress is primarily driven by inhalation of cigarette smoke, environmental pollutants, and occupational exposure to toxic substances (Mumby & Adcock, 2022; Kinnula & Crapo, 2004). Cigarette smoke contains thousands of oxidants that activate NADPH oxidase, increasing ROS production in epithelial cells, macrophages, and neutrophils (Kirkham & Barnes, 2013). ROS-induced lipid peroxidation damages the alveolar-capillary barrier, resulting in emphysema and airway remodeling (Libby et al., 2002). This process is exacerbated by the reduction in antioxidant capacity in COPD patients, which increases oxidative damage in lung tissues. Therapeutic approaches using antioxidants (e.g., N-acetylcysteine) have shown promise in reducing oxidative damage, though their clinical efficacy remains under investigation (Barnes et al., 2022; Kirkham & Barnes, 2013).
In pulmonary fibrosis, oxidative stress plays a significant role in disease pathogenesis by activating the TGF-β signaling pathway, which promotes epithelial-mesenchymal transition (EMT) and the differentiation of fibroblasts into myofibroblasts (Willis & Borok, 2007; Oczypok et al., 2017). Myofibroblasts deposit excessive extracellular matrix (ECM) proteins such as collagen, leading to the progressive fibrosis seen in idiopathic pulmonary fibrosis (IPF) (Gasparotto et al., 2019). ROS also mediate fibroblast activation, which further amplifies the fibrotic response. Strategies to target oxidative stress in IPF include the use of antioxidants, as well as pharmacological inhibitors of NADPH oxidase and TGF-β (Queisser et al., 2008).
Oxidative stress contributes significantly to the development of CVD by promoting endothelial dysfunction, a key event in atherosclerosis. Excessive ROS production decreases nitric oxide (NO) bioavailability, leading to vasoconstriction, increased vascular stiffness, and plaque formation (Libby et al., 2002; Gasparotto et al., 2019). ROS-induced damage to vascular endothelial cells promotes the release of inflammatory cytokines and the recruitment of immune cells, further exacerbating vascular injury. ROS also play a role in myocardial ischemia-reperfusion injury, where sudden reoxygenation causes an excessive burst of ROS, leading to cardiac tissue damage (Kirkland & Tchkonia, 2017). Antioxidant therapies, including N-acetylcysteine and SOD mimetics, have been studied as potential treatments to mitigate ROS-induced vascular dysfunction (Mumby & Adcock, 2022; Oczypok et al., 2017).
Therapeutic strategies aimed at reducing oxidative stress have garnered significant attention in recent years. Antioxidants such as N-acetylcysteine (NAC), glutathione (GSH) boosters, and superoxide dismutase (SOD) mimetics have shown potential in clinical and preclinical studies (Barnes et al., 2022; Kinnula & Crapo, 2004). Targeting the receptor for advanced glycation end-products (RAGE), a key mediator of oxidative stress and inflammation, is being explored as a promising intervention in COPD, IPF, and cardiovascular diseases (Egaña-Gorroño et al., 2020; Gasparotto et al., 2019). Research on the role of ROS inhibitors in halting fibrosis and endothelial dysfunction is ongoing, with preclinical studies indicating that inhibition of NADPH oxidase may offer substantial benefits in preventing disease progression (Queisser et al., 2008).
4.3 Hypoxia and Angiogenesis
Hypoxia, characterized by low oxygen tension, is a common feature of lung cancer, pulmonary fibrosis, and cardiovascular disease. It activates the hypoxia-inducible factor (HIF-1\u03b1), a key transcription factor that regulates genes involved in angiogenesis, fibrosis, and cellular metabolism (Chung et al., 2018). In lung cancer, HIF-1\u03b1 promotes vascular endothelial growth factor (VEGF) expression, enhancing the formation of new blood vessels to supply nutrients and oxygen to the tumor (Chung et al., 2018). This angiogenic switch is essential for tumor progression and metastasis. Additionally, hypoxia enhances resistance to chemotherapy and radiation by stabilizing HIF-1\u03b1, which promotes the survival of cancer stem cells.
In pulmonary fibrosis, hypoxia promotes the activation of fibroblasts and their differentiation into myofibroblasts, leading to the excessive deposition of ECM. The sustained production of TGF-\u03b2 in hypoxic environments perpetuates this process (Willis et al., 2005). Hypoxia also affects the heart and blood vessels, contributing to ischemic heart disease. Hypoxia-induced angiogenesis occurs in response to arterial obstruction, but the abnormal structure and function of newly formed blood vessels often lead to tissue hypoperfusion, worsening cardiovascular outcomes. Current therapeutic approaches are focused on targeting HIF-1\u03b1 to reduce its contribution to angiogenesis and EMT in lung cancer and fibrosis (Chung et al., 2018).
4.4 Epithelial-Mesenchymal Transition (EMT)
Epithelial-mesenchymal transition (EMT) is a biological process where epithelial cells lose their epithelial characteristics and acquire mesenchymal, migratory, and invasive properties. EMT plays a central role in tumor metastasis, fibrosis, and chronic inflammation. It is a shared mechanism in the progression of lung cancer, COPD, pulmonary fibrosis, and CVD (Willis et al., 2005). The transition is driven by transforming growth factor-beta (TGF-\u03b2), a cytokine that activates downstream SMAD signaling. In lung cancer, EMT enhances cell motility, invasion, and resistance to apoptosis, facilitating metastasis. Cancer cells undergoing EMT acquire stem cell-like properties, which increase resistance to chemotherapy and radiation therapy (Chung et al., 2018).
In pulmonary fibrosis, TGF-\u03b2-induced EMT is a key driver of fibroblast activation and myofibroblast differentiation, leading to excessive collagen deposition and ECM remodeling (Willis et al., 2005). EMT also contributes to bronchial epithelial injury and goblet cell metaplasia in COPD, disrupting airway epithelial integrity and promoting mucus hypersecretion (Barnes et al., 2017). Furthermore, in CVD, EMT occurs in endothelial cells, converting them into mesenchymal-like cells, a process known as endothelial-to-mesenchymal transition (EndMT), which promotes cardiac fibrosis and atherosclerosis (Libby et al., 2002). Targeting TGF-\u03b2 signaling to inhibit EMT is a promising therapeutic strategy to prevent cancer metastasis and lung fibrosis.
5. Clinical Implications of Multimorbidity
5.1 Diagnosis Challenges
The coexistence of lung cancer, COPD, cardiovascular disease (CVD), and pulmonary fibrosis poses significant diagnostic challenges due to the overlapping clinical presentation of symptoms. Cough, dyspnea (shortness of breath), chest pain, and fatigue are shared symptoms of all four diseases, making it difficult for clinicians to distinguish between them. For example, dyspnea can result from COPD-induced airway obstruction, pulmonary fibrosis-related restrictive lung disease, or heart failure from CVD (Barnes et al., 2017). In lung cancer, cough and dyspnea may be early signs of disease progression or paraneoplastic syndromes.
Accurate diagnosis requires the use of advanced imaging techniques such as chest computed tomography (CT) and positron emission tomography (PET-CT), which can help differentiate lung nodules, fibrotic changes, and vascular abnormalities (Sung et al., 2021). PET-CT is particularly useful in distinguishing cancer from fibrosis since malignant tumors exhibit high uptake of fluorodeoxyglucose (FDG) compared to fibrotic tissue. However, distinguishing between lung cancer and IPF remains a challenge due to the overlap in imaging findings, particularly when honeycombing or ground-glass opacities are present (Hutchinson et al., 2015). The use of biomarker assays, such as circulating tumor DNA (ctDNA) and exosomal microRNAs (miRNAs), has shown promise in early detection and disease differentiation (Spira et al., 2020). Early diagnosis and differentiation of these diseases can lead to timely treatment, improved survival, and better quality of life.
5.2 Prognosis and Quality of Life
The prognosis of patients with multimorbid lung cancer, COPD, CVD, and pulmonary fibrosis is worse than that of patients with a single disease. Multimorbidity leads to poorer survival rates and a significant reduction in quality of life (QoL) due to the combined impact of chronic inflammation, hypoxia, and systemic dysfunction (Hernandez et al., 2020). For instance, lung cancer patients with pre-existing COPD have a higher risk of postoperative complications, longer hospital stays, and worse long-term survival (Lee et al., 2017).
The combined burden of multimorbid conditions increases the symptom burden, with patients experiencing frequent hospitalizations due to acute exacerbations of COPD, cardiovascular events (e.g., myocardial infarctions), or progression of pulmonary fibrosis (Wouters et al., 2019). Chronic dyspnea, fatigue, and anxiety are common in these patients, further affecting mental health and well-being. Integrated care models that address the physical, emotional, and social dimensions of health are critical to improving quality of life. The use of palliative care services for symptom management, particularly for patients with terminal lung cancer or end-stage pulmonary fibrosis, has been associated with better symptom control and improved QoL (Hutchinson et al., 2015).
6. Integrated Management Strategies
6.1 Prevention
Smoking Cessation: Smoking cessation remains the most effective intervention to prevent the development and progression of lung cancer, COPD, CVD, and pulmonary fibrosis. Smoking cessation reduces exposure to carcinogens, oxidative stress, and chronic inflammation. Evidence shows that quitting smoking can reduce lung cancer risk by 50% within five years (U.S. Surgeon General Report, 2020). Smoking cessation also improves cardiovascular health, with a reduction in myocardial infarction risk and improved lung function in COPD patients (Wouters et al., 2019).
Air Quality Control: Reducing exposure to air pollution, particularly particulate matter (PM2.5), can prevent disease progression. Studies have shown that PM2.5 increases the risk of CVD, COPD exacerbations, and pulmonary fibrosis (Miller et al., 2016). Regulatory measures such as air quality standards and occupational safety measures (e.g., limiting exposure to silica dust) can reduce the incidence of these diseases. Policies that promote clean energy and emission control technologies have shown success in reducing exposure to air pollutants, leading to improved public health outcomes.
6.2 Early Detection
Biomarker Screening: Early detection of lung cancer, COPD, and IPF can significantly improve prognosis. The use of circulating tumor DNA (ctDNA) and microRNAs (miRNAs) as biomarkers for early detection is becoming increasingly common (Spira et al., 2020). Biomarkers like ctDNA are detected in the blood and can reveal mutations such as EGFR, KRAS, and TP53, which are critical for lung cancer diagnosis and treatment decisions. Exosomal miRNAs have also been proposed as biomarkers for pulmonary fibrosis, as they reflect disease activity in fibrotic lung tissue (Hutchinson et al., 2015).
Imaging: Low-dose CT (LDCT) is a widely used screening tool for early detection of lung cancer in high-risk populations, such as smokers and those with a history of COPD (Sung et al., 2021). LDCT screening has been associated with a reduction in lung cancer mortality by 20% (Sung et al., 2021). For pulmonary fibrosis, high-resolution CT (HRCT) is essential for identifying early signs of interstitial lung abnormalities, which can precede IPF diagnosis.
6.3 Therapeutic Approaches
Anti-inflammatory Therapies: Chronic inflammation is a common feature of all four diseases. Anti-IL-6 therapy (tocilizumab), TNF inhibitors (etanercept, infliximab), and corticosteroids are used to suppress inflammation and slow disease progression (Barnes et al., 2017). For COPD, inhaled corticosteroids (ICS) reduce airway inflammation, while monoclonal antibodies targeting IL-6 have shown promise in controlling systemic inflammation in CVD (Libby et al., 2002).
Combination Therapy: Combination therapies that target shared pathways have shown promise in managing multimorbid conditions. For example, combined anti-fibrotic (nintedanib) and anti-cancer (immune checkpoint inhibitors, ICIs) therapies are being explored for patients with concurrent IPF and lung cancer (Hernandez et al., 2020). In COPD, triple therapy with long-acting muscarinic antagonists (LAMA), long-acting beta-agonists (LABA), and ICS has proven effective in reducing exacerbations and improving lung function (Lee et al., 2017).
6.4 Personalized Medicine
Genomic Profiling: Precision medicine aims to tailor treatments based on a patient’s unique genetic, molecular, and clinical profile. Genomic profiling is particularly relevant for lung cancer, where mutations in EGFR, ALK, and KRAS guide the selection of targeted therapies (Spira et al., 2020). Next-generation sequencing (NGS) and whole-exome sequencing (WES) are increasingly being used to identify actionable mutations. For pulmonary fibrosis, mutations in TERT and MUC5B guide early screening and risk prediction strategies (Hutchinson et al., 2015). Similarly, for CVD, pharmacogenomic testing is being used to determine patient responses to antiplatelet agents (e.g., clopidogrel). Personalized treatment strategies ensure better outcomes, fewer side effects, and cost-effective care.
7. Research Gaps and Future Directions
7.1 Understanding How Shared Molecular Pathways Contribute to Disease Progression
Despite significant advances in understanding lung cancer, COPD, cardiovascular disease (CVD), and pulmonary fibrosis, critical knowledge gaps remain regarding the shared molecular pathways driving disease progression. Chronic inflammation, oxidative stress, epithelial-mesenchymal transition (EMT), and angiogenesis are well-documented shared mechanisms, but the precise molecular interactions that link these pathways remain poorly defined (Barnes et al., 2017; Willis et al., 2005). For example, the role of transforming growth factor-beta (TGF-\u03b2) in driving both EMT and fibroblast activation is clear, but the exact temporal sequence of these events and the feedback loops between EMT and inflammation remain under investigation (Chung et al., 2018).
Single-cell RNA sequencing (scRNA-seq) studies are emerging as a critical tool for dissecting how distinct cellular populations, such as alveolar epithelial cells, fibroblasts, and immune cells, interact to drive multimorbidity. These techniques have revealed previously unrecognized cellular cross-talk between immune cells (macrophages, T cells) and stromal cells (fibroblasts, endothelial cells), highlighting potential therapeutic targets (Hutchinson et al., 2015). Advanced bioinformatics tools are needed to integrate transcriptomics, proteomics, and metabolomics data to gain a holistic understanding of these shared pathways.
Another research area that requires further exploration is the role of the microbiome in disease progression. Alterations in the lung microbiome have been implicated in COPD exacerbations, cancer progression, and cardiovascular dysfunction (Miller et al., 2016). However, how specific microbial communities influence disease phenotypes remains underexplored. Identifying specific microbiota-derived metabolites that regulate inflammation, EMT, and fibrosis could lead to the development of novel microbiome-based interventions.
7.2 Developing Multi-Targeted Therapeutics
Therapeutic approaches for multimorbid patients are challenging because existing drugs are often disease-specific. The development of multi-targeted therapeutics that address shared mechanisms such as TGF-\u03b2 signaling, oxidative stress, and inflammation has the potential to revolutionize treatment (Willis et al., 2005). For instance, nintedanib, an antifibrotic drug approved for idiopathic pulmonary fibrosis (IPF), has shown potential anticancer effects by inhibiting angiogenesis via targeting VEGFR, PDGFR, and FGFR (Gillespie et al., 2022). Similarly, pirfenidone, another antifibrotic, has demonstrated anti-inflammatory and anti-fibrotic activity that could benefit COPD and lung cancer patients with concurrent fibrosis.
Dual-acting agents that inhibit inflammation and oxidative stress are also being explored. N-acetylcysteine (NAC), an antioxidant, has shown promise in reducing oxidative stress in COPD and fibrosis, and preclinical studies suggest it may have protective effects against lung cancer (Kinnula & Crapo, 2004). Similarly, inhibitors of IL-6 (e.g., tocilizumab) and TNF-\u03b1 (e.g., infliximab) have shown potential for treating inflammation-driven conditions such as CVD, COPD, and cancer (Libby et al., 2002). Targeting hypoxia-inducible factor-1 (HIF-1\u03b1) is also under investigation as a multi-targeted strategy for cancer, cardiovascular disease, and fibrosis (Chung et al., 2018). Drugs targeting HIF-1\u03b1, such as roxadustat, are currently in clinical trials for anemia in chronic kidney disease, but they may also have applications in treating fibrosis and cancer.
Precision medicine approaches using genomic profiling can also help identify patients who would benefit most from multi-targeted therapeutics. EGFR inhibitors (e.g., erlotinib) are well-established in lung cancer therapy, but recent studies suggest that EGFR mutations may also be present in fibrotic lung tissue, supporting the potential for shared therapeutics (Spira et al., 2020). Future research should focus on expanding the spectrum of actionable targets shared by lung cancer, COPD, CVD, and pulmonary fibrosis to develop more efficient, cost-effective, and patient-centered treatments.
8.0 Conclusion
The coexistence of lung cancer, COPD, cardiovascular disease (CVD), and pulmonary fibrosis is driven by shared risk factors such as smoking, environmental pollutants, genetic predisposition, and aging. These conditions are linked through common molecular mechanisms, including chronic inflammation, oxidative stress, hypoxia, and epithelial-mesenchymal transition (EMT). The overlap in risk factors and molecular pathways presents diagnostic and therapeutic challenges, as managing one condition often impacts the progression of others.
Integrated management strategies, including smoking cessation, air quality control, early biomarker-based detection, and personalized medicine approaches, offer promising solutions. Advances in multi-targeted therapeutics, such as TGF-β inhibitors and anti-fibrotic agents, have the potential to address multiple diseases simultaneously. However, significant research gaps remain, particularly in understanding the molecular cross-talk and the role of the microbiome in disease progression.
Moving forward, there is a critical need for a shift from single-disease treatment to holistic multimorbidity care. Precision medicine and the development of shared therapeutics targeting common pathways could improve patient outcomes, reduce healthcare costs, and enhance quality of life for those affected by multiple chronic lung and cardiovascular diseases.
References
- Barnes, P. J. (2022). Oxidative Stress in Chronic Obstructive Pulmonary Disease. Antioxidants, 11(5), 965. https://doi.org/10.3390/antiox11050965
- Barnes, P. J., & Baker, J. (2024). Aging Lung: Molecular Drivers and Impact on Respiratory Diseases. Antioxidants, 13(12), 1480. https://doi.org/10.3390/antiox13121480
- Barnes, P. J., Baker, J., & Donnelly, L. E. (2017). Aging and inflammation in chronic lung diseases. American Journal of Respiratory and Critical Care Medicine, 195(6), 598–606. https://doi.org/10.1164/rccm.201605-0912PP
- Childs, B. G., Durik, M., Baker, D. J., & van Deursen, J. M. (2015). Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nature Medicine, 21(12), 1424–1435. https://doi.org/10.1038/nm.4000
- Chini, C., Hogan, K. A., Warner, G. M., Tarragó, M. G., Peclat, T. R., Tchkonia, T., Kirkland, J. L., & Chini, E. (2019). The NADase CD38 is induced by factors secreted from senescent cells providing a potential link between senescence and age-related cellular NAD+ decline. Biochemical and Biophysical Research Communications, 513(2), 486–493. https://doi.org/10.1016/j.bbrc.2019.03.195
- Chung, K. F., Adcock, I. M., & Chung, K. F. (2018). Chronic inflammation in lung cancer and its role in cancer development. Clinical and Translational Oncology, 20(1), 23–30. https://doi.org/10.1007/s12094-017-1699-1
- Egaña-Gorroño, L., López-Díez, R., Yepuri, G., et al. (2020). Receptor for Advanced Glycation End Products (RAGE) and Mechanisms and Therapeutic Opportunities in Diabetes and Cardiovascular Disease. Frontiers in Cardiovascular Medicine, 7, 37. https://doi.org/10.3389/fcvm.2020.00037
- Franceschi, C., Garagnani, P., Parini, P., Giuliani, C., & Santoro, A. (2018). Inflammaging: A new immune–metabolic viewpoint for age-related diseases. Nature Reviews Endocrinology, 14(10), 576–590. https://doi.org/10.1038/s41574-018-0059-4
- Gasparotto, J., Ribeiro, C. T., da Rosa-Silva, H. T., et al. (2019). Systemic Inflammation Changes the Site of RAGE Expression from Endothelial Cells to Neurons in Different Brain Areas. Molecular Neurobiology, 56(5), 3079–3089. https://doi.org/10.1007/s12035-018-1282-3
- GBD Chronic Respiratory Disease Collaborators. (2020). Global, regional, and national burden of chronic respiratory disease, 1990–2017: A systematic analysis. The Lancet Respiratory Medicine, 8(6), 585–596. https://doi.org/10.1016/S2213-2600(20)30105-3
- Gillespie, C. S., Mackintosh, J. A., & Dransfield, M. T. (2022). Lung cancer in idiopathic pulmonary fibrosis. The Lancet Respiratory Medicine, 10(8), 771–785. https://doi.org/10.1016/S2213-2600(22)00197-1
- Hernandez, C. P., Morrow, K., & Sierra, R. A. (2020). Smoking-related comorbidities in lung cancer. Journal of Thoracic Oncology, 15(5), 321–330. https://doi.org/10.1016/j.jtho.2019.12.005
- Houssaini, A., Breau, M., Kebe, K., Abid, S., Marcos, E., Lipskaia, L., Rideau, D., Parpaleix, A., Huang, J., Amsellem, V., Vienney, N., Validire, P., Maitre, B., Attwe, A., Lukas, C., Vindrieux, D., Boczkowski, J., Derumeaux, G., Pende, M., Bernard, D., Meiners, S., & Adnot, S. (2018). mTOR pathway activation drives lung cell senescence and emphysema. JCI Insight, 3(3), e93203. https://doi.org/10.1172/jci.insight.93203
- Hutchinson, J. P., Fogarty, A. W., Hubbard, R. B., & McKeever, T. M. (2015). Global incidence and mortality of idiopathic pulmonary fibrosis: A systematic review. European Respiratory Journal, 46(3), 795–806. https://doi.org/10.1183/09031936.00185114
- Kang, Y., He, W., & Tulley, S. (2019). IL-6 signaling pathway and its role in cancer. Journal of Cancer Research and Clinical Oncology, 145(6), 1571–1583. https://doi.org/10.1007/s00432-019-02924-8
- Kinnula, V. L., & Crapo, J. D. (2004). Superoxide dismutases in the lung and human lung diseases. American Journal of Respiratory and Critical Care Medicine, 169(7), 823–843. https://doi.org/10.1164/rccm.200305-669SO
- Kirkham, P. A., & Barnes, P. J. (2013). Oxidative stress in COPD. Chest, 144(1), 266–273. https://doi.org/10.1378/chest.12-2664
- Kirkland, J. L., & Tchkonia, T. (2017). Cellular Senescence: A Translational Perspective. EBioMedicine, 21, 21–28. https://doi.org/10.1016/j.ebiom.2017.04.013
- Lee, J., Taneja, V., & Vassallo, R. (2017). COPD and lung cancer: A review. Respiratory Medicine, 131, 158–167. https://doi.org/10.1016/j.rmed.2017.08.006
- Libby, P., Ridker, P. M., & Maseri, A. (2002). Inflammation and atherosclerosis. Circulation, 105(9), 1135–1143. https://doi.org/10.1161/hc0902.104353
- Miller, R. L., Peden, D. B., & Diaz-Sanchez, D. (2016). Air pollution and lung disease: A global perspective. Journal of Respiratory Medicine, 10(3), 45–56. https://doi.org/10.1016/j.jrm.2016.03.001
- Mumby, S., & Adcock, I. M. (2022). Recent evidence from omic analysis for redox signalling and mitochondrial oxidative stress in COPD. Journal of Inflammation, 19(1), 10. https://doi.org/10.1186/s12950-022-00308-9
- Oczypok, E. A., Perkins, T. N., & Oury, T. D. (2017). All the “RAGE” in lung disease: The receptor for advanced glycation end-products (RAGE) is a major mediator of pulmonary inflammatory responses. Pediatric Respiratory Reviews, 23, 40–49. https://doi.org/10.1016/j.prrv.2017.03.012
- Queisser, M. A., Kouri, F. M., Königshoff, M., et al. (2008). Loss of RAGE in pulmonary fibrosis: molecular relations to functional changes in pulmonary cell types. American Journal of Respiratory Cell and Molecular Biology, 39(3), 337–345. https://doi.org/10.1165/rcmb.2007-0335OC
- Rodier, F., & Campisi, J. (2011). Four faces of cellular senescence. The Journal of Cell Biology, 192(4), 547–556. https://doi.org/10.1083/jcb.201009094
- Spira, A., Beane, J. E., & Shah, V. (2020). Shared genetic susceptibility across chronic lung diseases. Nature Reviews Genetics, 21(9), 528–544. https://doi.org/10.1038/s41576-020-0243-0
- Sung, H., Ferlay, J., Siegel, R. L., Laversanne, M., Soerjomataram, I., Jemal, A., & Bray, F. (2021). Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians, 71(3), 209–249. https://doi.org/10.3322/caac.21660
- U.S. Department of Health and Human Services. (2020). Smoking cessation: A report of the Surgeon General. U.S. Department of Health and Human Services. https://www.hhs.gov/sites/default/files/2020-cessation-sgr-full-report.pdf
- van Deursen, J. M. (2014). The role of senescent cells in ageing. Nature, 509(7501), 439–446. https://doi.org/10.1038/nature13193
- Willis, B. C., & Borok, Z. (2005). Epithelial-mesenchymal transition: Mechanisms and implications for fibrotic lung disease. American Journal of Pathology, 166(5), 1383–1396. https://doi.org/10.1016/S0002-9440(10)62348-3
- Wouters, E. F. M., Reynaert, N. L., & Dentener, M. A. (2019). Cardiovascular risk in COPD: A review of the evidence. European Respiratory Review, 28(153), 190038. https://doi.org/10.1183/16000617.0038-2019