Surien O, Ghazali A. R, Masre S. F. Lung Cancers and the Roles of Natural Compounds as Potential Chemotherapeutic and Chemopreventive Agents. Biomed Pharmacol J 2019;12(1).
Manuscript received on :25-Jan-2019
Manuscript accepted on :04-Mar-2019
Published online on: 18-03-2019
Plagiarism Check: Yes
Reviewed by: Sadegh Rajabi
Second Review by: Ankur Singh Bist
Final Approval by: Prof. Juei-Tang Cheng

How to Cite    |   Publication History
Views Views: (Visited 1,573 times, 1 visits today)   Downloads PDF Downloads: 920

Omchit Surien, Ahmad Rohi Ghazali and Siti Fathiah Masre*

Biomedical Science Programme, Centre of Health and Applied Sciences, Faculty of Health Sciences, National University of Malaysia (UKM), 50300 Kuala Lumpur, Malaysia.

Corresponding Author E-mail: sitifathiah@ukm.edu.my

DOI : https://dx.doi.org/10.13005/bpj/1617

Abstract

Among all types of human cancers, lung cancer is one of the most common and has the highest mortality rate. Two major groups of lung cancer based on histological features are non-small cell lung cancers (NSCLCs) and small cell lung cancers (SCLCs). NSCLCs are further subdivided into few subtypes such as adenocarcinoma (AD), squamous cell carcinoma (SCC) and large cell carcinoma(LCC). Focusing on specific treatments or molecular targeted therapy for each type of lung cancers promising the better results as each subtype of lung cancers differ in genomic alterations, the cell of origin and growth pattern. Numerous studies have been done to search for the best chemotherapeutic and chemopreventive agents that can reduce the burden of lung cancer, primarily focusing on potential natural products. This review is focused on genomic alterations and current potential natural compounds as chemotherapeutic and chemoprevention agents specifically on each subtype of lung cancers.

Keywords

Chemotherapeutic; Chemoprevention; Lung Cancers; Non-Small Cell Lung Cancers; Natural Compounds; Small Cell Lung Cancer

Download this article as: 
Copy the following to cite this article:

Surien O, Ghazali A. R, Masre S. F. Lung Cancers and the Roles of Natural Compounds as Potential Chemotherapeutic and Chemopreventive Agents. Biomed Pharmacol J 2019;12(1).

Copy the following to cite this URL:

Surien O, Ghazali A. R, Masre S. F. Lung Cancers and the Roles of Natural Compounds as Potential Chemotherapeutic and Chemopreventive Agents. Biomed Pharmacol J 2019;12(1). Available from: https://bit.ly/2TJxWG2

Introduction

The statistics reported by the World Health Organization (WHO) showed that lung cancer (including trachea and bronchus cancer) is one of the leading cause of deaths worldwide, causing about 1.7 million deaths in 2015.1 Lung cancer can be due to inborn genetic defects, environmental and lifestyle factors. While inborn genetic defects have little contribution, both environmental and lifestyle factors play a pivotal role in causing lung cancer.2,3 Asbestos, ionizing radiation, sulfur mustard, coal-tar pitch and tobacco smoking are some examples of environmental and lifestyle factors that serve as lung carcinogens.4 Tobacco smoking has been reported as the most influential risk factor for lung cancer cases in 90% of men and 80% of women.5 According to the International Agency for Research on Cancer, cigarette smoke contains 4000 identifiable chemicals with more than 60 chemicals are carcinogens.6 Polycyclic aromatic hydrocarbons (PAHs), N-nitrosamine, and aromatic amines are some of the most potent carcinogens that can be found in the cigarette smoke.7 Based on histological observation under the light microscopy, the two major groups of lung cancer are small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). NSCLC is further sub-divided into adenocarcinoma, squamous cell carcinoma and large cell carcinoma and others.8,9 NSCLC is more dominant and makes up almost 85% of all lung cancers while SCLC counts for about 15% of all lung cancers.10 The most common subtypes of NSCLC are lung adenocarcinoma and lung squamous cell carcinoma (SCC).11,12

The high mortality rate of lung cancer patients is due to the late diagnosis, with 70% of lung cancer cases identified in advanced stages.13 Late diagnosis has led to the failure of cancer treatment such as chemotherapy and radiotherapy.14 Early detection of lung cancer can substantially increase the chances of survival, where 70-90% of lung cancer patients with NSCLCs in early stage (stage I) that have been through surgical resection procedure can survive for five years.15,16 Difficulty in the detection of an early stage of lung cancer, high cost in diagnostic tests, and invasive diagnostic procedures have been linked to the late diagnosis of lung cancer with most patients were diagnosed with stage IIIB and IV.17 A population-based study also showed that approximately 75% of lung cancer patients were diagnosed at the advanced stages such as stage III and IV.18 Tables 1 provides the information about the latest 8th TNM staging system for NSCLCs and the explanation for each stage that differentiate based on three characteristics including the primary tumour, involvement of lymph node and state of metastasis.19

Table 1: 8th edition of the American Joint Commission on Cancer TNM staging system for non–small cell lung cancer(NSCLC) and the explanation for abbreviation of each stage (19).

Staging   TNM system (T: Primary tumor; N: Lymph node; M: Distant metastasis).
IA1 N0 with any T1a
IA2 -N0 with T1b
IA3 -N0 with T1c
IB -N0 with T2a
11A -N0 with T2b
11B -N0 with T3

-N1 with any T1a, T1b, T1c, T2a, or T2b.

IIIA -N0 with T4.

-N1 with any T3 or T4

-N2 with any T1a, T1b, T1c, T2a, or T2b.

 IIIB -N2 with any T3 or T4.

-N3 with any T1a, T1b, T1c, T2a, or T2b.

 IIIC -N3 with any T3 or T4
 IVA -Any N0 to N3 with any M1a or M1b
IVB -Any N0 to N3 with M1c
Explanation
1) Primary tumour(T)
T1a Tumour in central airway but only superficial spreading (tumor equal or less than 1cm).
T1b Tumour more than 1 cm but less than or equal to 2cm.
T1c Tumour more than 2 cm but less than or equal to 3cm.
T2a Involvement of visceral pleura, main bronchus, atelectasis to hilum, and no carina involvement. Tumour more than 3cm but less than or equal to 4cm.
T2b Involvement of visceral pleura, main bronchus, atelectasis to hilum, and no carina involvement. Tumour more than 4 cm but less than or equal to 5cm.
T3 Invasion of tumour to the chest wall, pericardium, phrenic nerve; or one or more tumor nodules in same lobe with primary tumour; or tumour more than 5 cm but less than or equal to 7cm.
T4 Invasion of tumour to the mediastinum, diaphragm, heart, great vessels, recurrent laryngeal nerve, carina, trachea, esophagus, and spine; or tumor nodules in different ipsilateral lobe from where the primary tumor located; or tumour more than 7cm.
2)Lymph node(N)
N0 No of regional lymph node metastasis.
N1 Involvement of primary tumour that metastasis to  the ipsilateral pulmonary or hilar lymph nodes.
N2 Involvement of primary tumour that metastasis to the mediastinal or subcarinal lymph nodes.
N3 Involvement of primary tumour that metastasis to the contralateral mediastinal, hilar or supraclavicular lymph nodes.
3) Distant metastasis(M)
M1a Metastasis of primary tumour to the pleural or pericardial; or  metastasis of tumor to the contralateral lobe from primary tumor.
M1b Metastasis of tumour to any single extrathoraric.
M1c Metastasis of tumour to multiple extrathoraric (1 or more organs).

 

Treatments

The current treatment for lung cancer includes surgery, adjuvant, chemotherapy, and radiotherapy. The treatment of lung cancer depends on the histology and tumour stage. For example, in NSCLC, surgery will be done in patients with stage I, II, and IIIa if a tumour is resectable and depends on patient’s toleration toward surgery.20 However, patients who have stage IIA, IIB, and IIIA NSCLC need subsequent treatment after the surgical procedure such as chemotherapy.21 Differently, the first line treatment for SCLC patients is chemotherapy (combination of platinum and etoposide) as surgical resection is rarely done due to early metastasis in this type of lung cancer. SCLC is very sensitive to chemotherapy during the initial treatment, but most of the SCLC patients (up to 90%) tend to develop chemoresistance. Besides, chemotherapy is also the only therapeutic option for the patients with metastatic NSCLC though most of the patients die as they cannot be treated through surgery procedure.22

The only curative treatment for lung cancer is a radical surgery,23 yet, delay in diagnosis will lead to the poor prognosis. Unfortunately, 40% of the newly diagnosed NSCLC cases were advanced to stage IV and surgery alone is not enough for the treatment of lung cancer patients in the late stage. Cytotoxic chemotherapy is the first line therapy for NSCLC patients with stage IV that includes four to six cycles of platinum-based therapy such as cisplatin and carboplatin, with maintenance therapy afterwards in some patients is needed.24-26 There have been many reports on several complications, and side effects of conventional treatment approaches including the development of drug resistance in cancer cells due to the application of synthetic chemotherapeutic agents and the cost for synthetic drugs itself in cancer treatment is more expensive.20,27 Furthermore, most of the drugs that used in current cancer treatments have many disadvantages such as rapid metabolism, non-selective, irritating nature, low aqueous stability, and others that consequently lead to the several adverse effects such as low quality of life in cancer patients and dose-limiting side effects.28 Alternative strategies needed to control this disease and there are many aspects should be improved in lung cancer treatments such as chemotherapy due to insignificant improvement in the efficacy of treatment in recent decades.29

Chemoprevention

Sporn first introduced the term of chemoprevention in the early 1970s and defined it as the process of inhibition, suppression or reversion of cancer development or progression by using any natural or synthetic agents.30 Carcinogenesis is a complex event, and its development is a multistage process that consists of several phases: initiation, promotion, progression and lastly metastasis.31 For example, lung carcinogenesis derives from multiple steps involving a series of genetic and epigenetic alterations in pulmonary epithelial cells that lead to the changes in cell proliferation, differentiation, invasion, and metastasis.32

Moreover, lung carcinogenesis also showed the characteristic of “field of cancerization” that refers to the adjacent tissue to neoplastic lesions that appear histologically normal, but this tissue consists of molecular abnormalities as in the tumours.33 Example of the “field of cancerization” during lung carcinogenesis is the formation of premalignant or injury in all airway of epithelial cells after exposure to the cigarette smoke.34,35 This concept is further supported because of a single mutant epithelial cell in respiratory lining potentially to expand and extend into surrounding tissues in lung and lastly turns into malignancy. Even after smoking cessation, the expansion of premalignant field can still occur, and it is the vital step in lung carcinogenesis.36 Therefore, effective chemoprevention strategies are urgently needed as lung cancer risk is persistently increased even after the smoking cessation (former smokers).37

Natural agents such as phytochemicals are selectively targeting cancer cells, and even in small doses amount, the chemopreventive phytochemicals regularly act on cancer cells without affecting healthy cells.38 The natural agents from fruits and vegetables have shown several chemopreventive potentials in lung cancer. For example, one study revealed that the consumption of fruits and vegetables in current smokers could reduce the risk of getting lung cancer.39 Moreover, another study also showed the same finding where the higher consumption of cruciferous vegetables together with cigarette smoking control could lower the risk of lung cancer.40 Accordingly, there must be many potential candidates from natural sources that can be developed as cancer therapeutic and preventive agents to reduce the adverse effects of conventional cancer treatment.

Non-Small Cell Lung Cancer (NSCLC)

Lung Adenocarcinoma (AD)

Lung AD is a type of cancer that develops or arises from epithelial that line the small peripheral airways and categorised under the subtype of non-small cell lung cancer (NSLC). It is the most common type of all lung cancers41,42; hence lung adenocarcinoma is the most studied among other lung cancers. Amid all types of lung cancer, the incidence rate of lung adenocarcinoma in women is rapidly increased, but it is stabilised in men.43 Lung adenocarcinoma has a poor prognosis due to late diagnosis usually at the metastatic stage leading to failure in treatment.44 Although lung AD is the most frequent type of lung cancer seen in non-smokers, it also occurs in smoker patients.45 Other than that, in a multiracial Asian country, lung adenocarcinoma is more common in younger patients below the age of 40 years old compared to the older patients that have never smoked.46

The precursor of lung AD involves various genetic changes with the most common are p53 mutation (50% to 70%), epidermal growth factor receptor (EGFR) kinase domain mutation (10% to 40%), Kristen Rat Sarcoma viral oncogene (KRAS) mutation (10% to 30%), and LBK1 mutation (34%).47,48 Both KRAS and EGFR mutations are rarely found in one tumour.49 KRAS mutation is more common in smokers with an early stage of lung adenocarcinoma or atypical adenomatous hyperplasia (AAH), and EGFR kinase domain mutation is much more common in nonsmokers with lung adenocarcinoma.47,50  Moreover, 78% of lung AD patients had EGFR kinase domain mutation, but only 1.9% of patients had KRAS mutations among East Asian nonsmokers. EGFR plays various roles in a tumourigenic process including proliferation, apoptosis, angiogenesis and invasion.47

Molecular targeted therapy such as targeted kinase inhibitors by gefitinib (Iressa) was performed in lung adenocarcinoma treatment, and this drug has been approved in Japan and the United States.51 In contrast, there is still no treatment targeting the KRAS mutations. As one of the most common genetic defects in lung AD, KRAS mutations can be the potential genetic abnormality to be targeted in the treatment of lung AD.52 There have been are many studies reported on the potential of natural agents to give effects as anti-cancer and chemoprevention on lung adenocarcinoma with various underlying mechanisms. Table 2 shows the summary of chemoprevention and chemotherapeutic recent researches on natural products against lung AD including the mechanisms of action.

Table 2: The summary of chemoprevention and chemotherapeutic effects of natural products against lung AD (53-64).

Natural product Sources Mechanisms of action Reference
Chemoprevention 
Pterostilbene

 

Vitis vinifera leaves and fruits; blueberries and cranberries In vivo:

Pterostilbene decreased the epidermal growth factor receptor EGFR expression and also decreased the expression of other EGFR mediators such as Akt/mTOR, ERK1/2, Stat-3, and NFкB.

Pterostilbene also induced apoptosis by decreasing the level of caspase-3 and LC3-II.

Chen et al. 2012 (53).
β-escin Horse chestnut seeds (Aesculus hippocastanum). In vivo:

Reduced the number of tumours in a mouse model. β-escin induced cell cycles arrest by reducing the expression of ALDH1A1, p-Akt, RhoA, and ROCK proteins. However, it increased the expression of p21 protein and reduced the level of PCNA.

Patlolla et al. 2013(54).
Cucurbitacin B (CuB) Cucurbitaceae plants such as fruit of L.graveolense Roxb. In vitro:

CuB induced cell cycle arrest at G2/M phase and apoptosis.

In vivo:

Reduced the tumour multiplicity and PCNA-positive cells.

In vitro and in vivo:

Upregulation of p16 and p21 proteins expression and downregulation of tumour suppressor proteins such as c-myc, KRAS, and hTERT.

CuB inhibited the expression of DNMTs and histone deacetylase (HDACs).

Shukla et al. 2015 (55).
Chemotherapeutic
Curcumin Rhizome of Curcuma

longa.

In vitro:

Curcumin treatment inhibited human lung adenocarcinoma derived cells (H441 cells) by suppressing the activation of the Stat-3 pathway.

Alexandrow et al. 2012 (56).
Honokiol Magnolia grandiflora. In vitro:

Inhibited cell growth and induced cell cycle arrest at the G1 phase of A549 and H1299 cell lines.

Repressed histone deacetylase (HDAC) activity and class 1 HDACs protein expression, but increased the activity of HATs.

In vivo:

Inhibited tumor growth of A549 and H1299 cell lines in a xenograft model. Induced apoptosis by downregulating Bcl-2 and Bcl-xl proteins expression.

The levels of class I HDACs and HDACs activity also decreased in this in vivo study.

Singh et al. 2012 (57).
Emodin

 

Roots and rhizomes Rheum palmatum L. In vitro:

Emodin suppressed the proliferation of A549 cell lines by activation of  ERK1/2 and downregulation of ERCC1 and Rad51  proteins expression that led to cytotoxicity.

He et al. 2012 (58).
Diosein The roots of Polygonatum zanlanscianense Pamp. In vitro:

Inhibited proliferation of A549 cell lines by causing DNA damage and cell cycle arrest at S phase.

Diosein induced apoptosis by upregulating the pro-apoptotic proteins expression (Bax, Bak, and Bid). Downregulated the anti-apoptotic proteins expression (Bcl-2 and Bcl-xl). Increased the caspase-3 and caspase-6 activities.

Wei et al. 2013 (59).
Piperine Black pepper (Piper nigrum) and long pepper (Piper longum). In vitro:

 Piperine inhibited the proliferation of A549 cell lines by causing cell cycle arrest at the G2/M phase via p53 dependent mitochondrial signalling pathway.

Induced apoptosis by activating caspase-3 and caspase-9, increased BAX and p53 proteins expression and downregulated Bcl-2 protein expression.

Lin et al. 2014 (60).
Plumbagin (PL) Plumbago

zeylanica L.

In vitro:

Inhibited proliferation of A549 and H23 cell lines by causing apoptosis and cell cycle arrest at G2/M phase.

Cell cycle arrest by downregulating the expression of cyclin B1 and Cdc2, but upregulated the expression of p53 and p21.

Induction of apoptosis by downregulating the expression of Bcl-2 and upregulating the expression of Bax and cytochrome c. Increased the levels of cleaved caspase-3 and 9.

PL also increased the reactive oxygen species (ROS) production.

Inhibited PI3K/Akt/mTOR pathway to induce autophagy as well.

Li et al. 2014 (61).
TXA9 The roots of Streptocaulon juventas. In vitro:

Induced apoptosis by increasing the level of Fas protein, Fas-associated death domain (FADD) and enzymes caspase-3 and caspase-8.

In vivo:

Inhibited tumour growth in a xenograft model.

Xue et al. 2015 (62).
Epigallocatechin-3-gallate (EGCG) Green tea In vitro:

EGCG inhibited nicotine-induced migration and invasion of A549 cells by suppressing the level of HIF-1α, vascular endothelial growth factor (VEGF), COX-2, p-Akt, p-ERK and vimentin proteins expression.

EGCG also downregulated expression of p53 and β-catenin proteins.

Shi et al. 2015 (63).
Picropodophyllin. Mayapple plant family (Podophyllum peltatum). In vitro:

Inhibited proliferation of A549 and H1299 cell lines by repressing the expression of insulin-like growth factor 1 (IGF-1R) and decreasing the  p-Akt and MAPK. Induced apoptosis by increasing the level of caspase 3, 7 and PARP proteins.

Zhang et al. 2015 (64).

 

*EGFR= epidermal growth factor receptor; Akt/mTOR = protein kinase B/mammalian target of rapamycin (mTOR); ERK 1/2= extracellular-signal-regulated kinase; ALDH1A1= aldehyde dehydrogenase 1A1= ALDH1A1; Stat-3= signal transducer and activator of transcription3; LC3-II= microtubule-associated proteins 1A/1B light chain 3B-11; p-AKT= phospho-Akt; RhoA= Ras homolog gene family member A; ROCK= Rho-associated protein kinase; p21= cyclin-dependent kinase inhibitor 1; hTERT= human telomerase reverse transcriptase; KRAS= Kristen rat sarcoma 2 viral oncogene homolog; DNMTs= DNA methyltransferase; HDACs= histone deactylases; Stat-3= signal transducer and activator of -3; HDAC= histone deacetylase; HATs= histone acetyltransferases;Bcl-2= B-cell lymphoma 2; Bcl-xl= B-cell lymphoma-extra large; ERK 1/2= extracellular-signal-regulated kinase; ERCC1=excision repair cross-complementation group 1; RAD51= RAD51 recombinase;  BAX=bcl-2-like protein 4; Cdc2=cyclin-dependent kinase 1; PI3K=phosphoionositide 3-kinase;FADD=Fas-associated protein with death domain; HIF-1α=hypoxia inducible  factor 1 alpha; VEGF=vascular endothelial growth factor ; COX-2= cyclooxygenase-2; MAPK=mitogen activated protein kinases; PARP= poly(ADP-ribose) polymerase.

Squamous Cell Carcinoma (SCC)

Lung squamous cell carcinoma is an abnormal growth of cells from the bronchial epithelial cells through hyperplasia/metaplasia with the most common features are keratin pearls and/or intercellular bridges.65 The development of lung SCC is due to inhaled carcinogens that directly expose to the respiratory epithelium, and it is a sequential process that starts with squamous metaplasia, dysplasia, and carcinoma in situ.66 Historically, lung SCC is the most common type of lung cancer, but recently the lung adenocarcinoma has replaced the lung SCC as the most common type of lung cancer.12 Even though the reasons behind this trend is unknown, yet there are several possible factors such as diagnostic advances, changing from high-tar to low-tar filtered cigarettes and variations in smoking patterns.67 Cigarette smoking has a connection with all histologic types of lung cancer. However, a study done in 2001 revealed that smoking associated strongly with the lung SCC cases as compared to the carcinogenesis of lung adenocarcinoma.68 This strong association of smoking and lung SCC also has been proven by another study, where 96% of lung SCC patients in North America were ex-smokers or smokers.69

None of the targeted therapies for lung SCC has been approved due to the lack of genomic understanding. Moreover, genomic studies on lung SCC is only now emerging, unlike multiple targeted therapies that have been identified in lung adenocarcinoma.70 For example, the therapeutic approaches that targeted on EGFR and echinoderm microtubule-associated protein like 4 – anaplastic lymphoma kinase (EML4-ALK) are only applicable in lung adenocarcinoma treatment, but not in lung SCC.71 For example, a clinical study in 2006 reported that bevacizumab that acts as endothelial growth factor (VEGF) inhibitors together with standard treatment (platin-based chemotherapy) increased the survival of NSCLC patients. The Eastern Cooperative Oncology Group (ECOG) performance status of patients with advanced NSCLCs also better compared to the standard treatment alone without bevacizumab. Unfortunately, the results are different for lung SCC patients, where there is no improvement in survival and ECOG performance status in lung SCC patients.72 Thus, the treatment that effective for lung adenocarcinoma does not promise the same results in lung SCC. More studies on lung SCC is needed due to the limited progression in lung SCC treatment as compared to lung adenocarcinoma, even though both are the most common types of lung cancer.

The most common genetic changes in lung SCC is the mutation of TP53 gene with 60 to 70% of lung SCC cases had this mutation, followed by phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) amplification that accounted for 33% of lung SCC cases.47 Apart from the alterations of TP53 and PIK3CA, Cancer Genome Atlas Research Networks 2012 revealed the list of most common genetic alterations in lung SCC including the mutations of histone-lysine N-methyltransferase 2D(MLL-2), cyclin-dependent kinase 2A (CDKN2A), nuclear factor erythroid 2 like2 (NFE2L2), Kelch-like ECH-associated protein 1(KEAP1), and others. Others common genetic changes or alterations in lung AD such as mutations of EGFR and KRAS and fusion of anaplastic lymphoma kinase(ALK), but all these types of genetic changes are infrequent in lung SCC cases.71 Table 3 shows the the summary of chemoprevention and chemotherapeutic recent researches on natural products against lung SCC including the mechanisms of action.

Table 3: The summary of chemoprevention and chemotherapeutic effects of natural products against lung SCC (58, 64, 73-76).

Natural products  Sources Mechanisms of action Reference
Chemoprevention
Honokiol Bark of Magnolia tree. In vivo:

Honokiol interrupted the mitochondrial function by causing inhibition of mitochondrial respiration leading to a decreased in ATP levels. The decreased in ATP levels activated the AMPK  and lastly inhibited the tumour growth.

Honokiol enhances the ROS production in mitochondria.

Induction of  apoptosis by increasing the level of cleaved caspase-3.

Pan et al. 2014(73).
Picropodophyllin. Mayapple plant family (Podophyllum peltatum). In vivo:

Reduced the tumour load and multiplicity in benzo(a)pyrene-induced lung tumours in a mouse model by increasing the levels of cleaved caspase-3 apoptotic protein.

Zhang et al. 2015 (64).
Vitamin D3 In vivo:

Vitamin D3 inhibited the proliferation rate by reducing the levels of Ki67 protein.

Vitamin D3 induced the anti-inflammatory activity by reducing the expression of IL-6, and the white blood cell (WBC) counts.

Mazzilli et al. 2015 (74).
Chemotherapeutic
Emodin Roots and rhizomes Rheum palmatum L. In vitro:

Emodin inhibited the proliferation of SK-MES-1 cell lines (human lung squamous carcinoma cells). Emodin activated the ERK1/2 and downregulated the expression of ERCC1 and Rad51 proteins that caused cytotoxicity.

He et al. 2012 (58).
Alantolactone Inula helenium L. roots. In vitro:

Inhibited SK-MES-1 cells growth by causing apoptosis and cell cycle arrest at the G0/G1 phase.

Apoptosis- Downregulated the expression of Bcl-2, procaspase 9 and 3. Upregulated the expression of  Bax, cleaved caspase-3, and PARP.

Cell cycle arrest- Downregulated the expression of CDK4, CDK6, cyclin D3, and cyclin D1. Upregulated the expression of p21 and p27.

Zhao et al. 2015 (75).
Custonolide Compositae and  Magnoliaceae plant families. In vitro:

Induced cell cycle arrest at the G1/S phase and apoptosis in SK-MES-1 cell lines.

Apoptosis by downregulating Bcl-2 and procaspase-3 expression and upregulating Bax and cleaved PARP proteins expression.

Caused cell cycle arrest by upregulating p53, p21, and p27 expression, and downregulating pRB proteins.

Hua et al. 2016 (76).

 

*ATP= adenosine triphosphate; AMPK= 5’ AMP-activated protein kinase; Ki-67cell proliferation antigen Ki-67; IL-6 = interleukin-6; ROS=reactive oxygen species. ERK1/2=extracellular signal-regulated kinases 1/2; Rad51=DNA repair protein Rad51;ERCC1=DNA excision repair protein ERCC-1; CDK4=cyclin dependent kinase -4; CDK6; cyclin dependent kinase-6;pRB=phosho-retinoblastoma protein; p21=cyclin dependent kinase inhibitor 1; p53=tumor suppressor p53; p27cyclin dependent kinase inhibitor 1B.

Large Cell Carcinoma (LCC)

Large cell lung carcinoma (LCC) is a poorly differentiated subtype that often arises in peripheral lung tissue.77 In 2013, a team of researchers revealed that the tumour site of lung LCC is mostly located at the lung periphery with 81 cases (67%) out of 121 casesalso showed that LCC cases have a strong relation with smoking habit in which about 92% of patients are smokers or former smokers.78

Large cell neuroendocrine carcinoma (LCNC) is one example of lung large cell carcinoma (LCC), that shares the same characteristics with small cell lung cancer (SCLC) such as massive proliferation rates and neuroendocrine phenotype but lacks cytomorphology of SCLC.79,80 LCNC is a rare type of lung cancer with estimated to account only for 3% of all lung cancers.81 The most common genetic changes in LCNC is the alteration of TP53 that accounts for 78% of LCNC cases. Others common genetic changes in LCNC including the alteration of few genes that encoded for retinoblastoma (RB1) protein, serine/threonine kinase II (STK II), Kelch-like ECH-associated protein 1 (KEAP1), and KRAS.80 Chemoprevention of lung LCC was excluded from the discussion due to limited chemopreventive study on lung LCC. Besides being a less common type of lung cancer, the limited chemopreventive study of lung LCC might be due to difficulties of developing the animal model that relevance to the human lung LCC. Table 4 shows the summary of chemotherapeutic researches of natural products against lung LCC and SCLC.

Table 4: The summary of chemotherapeutic researches of natural products against lung LCC and SCLC (59, 82, 83, 94-97).

Natural products Sources Mechanisms of action Reference
Lung large cell carcinoma (LCC)
Didymin Flavonoid glycoside from citrus fruits. In vitro:

Inhibited H460 cells growth by inducing apoptosis.

Induced apoptosis by downregulating the expression of Fas/Apo-1 receptor and Fas-ligand. Didymin also activated the caspase-8.

Hung et al. 2010 (82).
α-tomatine Lycopersicon

esculentum Linn.

In vitro:

α-tomatine showed anti-metastatic effects on NCI-H460 cell lines.

Reduced the mRNA level and MMP-7 protein expression.

α-tomatine suppressed degradation and phosphorylation of DNA-binding activity of NF-кB and FAK/PI3K/Akt signalling pathway.

Shieh et al. 2011 (83).
Diosein Polygonatum zanlanscianense Pamp In vitro:

Inhibited proliferation and induced apoptosis in NCI-H460 cells.

Inhibited cell growth by causing DNA damage and cell cycle arrest at the S phase.

Apoptosis: upregulated the expression of pro-apoptotic proteins (Bax, Bak, and Bid).

Downregulated the expression of anti-apoptotic proteins (Bcl-2 and Bcl-xl).

Increased the caspase-3 and caspase-6 activities.

Wei et al. 2013 (59).
Small cell lung cancer(SCLC)
Curcumin Rhizome of Curcuma

longa

In vitro:

Inhibited cell proliferation, migration and invasion of human small cell lung carcinoma (NCI-H446 cell line).

Curcumin downregulated the expression of STAT3 by inhibiting the IL-6 leading to the inhibition of STAT3 phosphorylation.

Induced cell cycle arrest at the G2/M phase by downregulating the expression of Survivin, Bcl-xL and Cyclin B1.

Depressed the migration and invasion of cells by suppressing the levels of VEGF, MMP-2 and MMP-7 and ICAM-1.

Yang et al. 2012 (94).
Catechins Green tea In vitro:

Caused cell cycle arrest at the G2/M phase of NCI-H466 cell lines.

Upregulated the expression of let-7a-1 and let-7g protein leading to the downregulation of C-MYC and LIN-28 expression and induced cell cycle arrest.

 

 

Zhong et al. 2012 (95).

Diosein The root of Polygonatum zanlanscianense Pamp In vitro:

Inhibited cell proliferation and induced apoptosis in NCI-H446 cells.

Anti-proliferation: induced DNA damage and cell cycle arrest at the S phase.

Apoptosis: upregulated the expression of pro-apoptotic proteins (Bax, Bak, and Bid). Downregulated the expression of anti-apoptotic proteins (Bcl-2 and Bcl-xl). Diosein also increased the caspase-3 and caspase-6 activities.

Wei et al. 2013 (59).
Wentilactone A(WA) Marine-derived endophytic fungi Aspergillus wentii.  In vitro:

Induced apoptosis and cell cycle arrest at the G2/M phase in NC1-H446 cell lines.

Induced cell cycle arrest by downregulating the expression of cyclin B1, cdc2, p-cdc2, cdc25C and p-cdc25C. WA increased the phosphorylation of ERK, JNK, p38 to activate the mitogen-activated protein(MAP) kinase signalling cascades. WA also induced the expression of p53, p21 and ROS production to induce cell cycle arrest.

Apoptosis- Downregulated the expression of Bcl-2 and Mcl-1, but upregulated the expression of Bax, Bad, cleaved caspase-2 and caspase 7.

Lv et al. 2013 (96).
Evodiamine Evodia rutaecarpa In vitro:

Induced cell cycle arrest at the G2/M phase and apoptosis in H446 and H1688 cell lines.

Increased the ROS production and activities of caspase 8, 9 and 3.

Increased the expression of cytochrome C, caspase 3, 8, 9, and Bax proteins, and decreased the expression of Bcl-2 protein.

Fang et al. 2014 (97).

 

*Apo-1= Apoliprotein A1; MMP-7= matrix metalloproteinase-7; NF-kβ= nuclear factor kappa-light-chain enhancer of activated B cells; FAK= focal adhension kinase; PI3K= phosphatidylinositol 3-kinase; Akt= protein kinase B; Bax= Bcl-2 associated X protein; Bak= Bcl-2 homolog antagonist/killer; Bid=BH3 interacting-domain death agonist; Bcl-2= B cell lymphoma 2; Bcl-xL=  B cell lymphoma extra large. STAT3= Signal transducer and activator of transcription 3; IL6+interleukin 6; VEGF=vascular endothelial growth factor; MMP=matrix metalloproteinase; ICAM-1= intracellular adhension molecule 1; LIN28= Lin-28 homolog A protein; cdc25c= M-phase inducer phosphatase 3; ERK= extracellular-signal regulated kinase; JNK= c-Jun N-terminal kinases; ROS= reactive oxygen species; Bax= Bcl-2 associated X protein; Bak= Bcl-2 homolog antagonist/killer; Bid=BH3 interacting-domain death agonist; Bcl-2= B cell lymphoma 2; Bcl-xL=  B cell lymphoma extra large; Bad; Bcl-2 –associated death promoter.

Small Cell Lung Cancer (SCLC)

Small cell lung cancer (SCLC) is a type of lung cancer that develops from neuroendocrine-cell precursors. Some essential characteristics of SCLC are rapid cell growth, highly vascularized, genomic instability and early onset of metastatic. SCLC has the lowest survival rate among all type of lung cancers with only 3% to 8% of five-year survival rate, but the five-year survival rate of other types of lung cancers are more than 15%.84 Most of the SCLC patients are diagnosed with metastatic as the cancerous cells disseminated rapidly through the blood and lymphatic system.85 The response rates of SCLC towards both chemotherapy and radiotherapy is high, yet, patients with metastatic SCLC tends to develop treatment resistance.86,87 The high risk of SCLC has a strong connection with cigarette smoking. Smoking cessation has an enormous impact on SCLC by reducing the risk of getting SCLC and increasing the survival rate among the patients with localised SCLC by almost 50%.88 Moreover, a study found that among 148 SCLC patients only 3 patients were non-smokers and another 145 patients were smokers.89

The most common molecular genetic changes in SCLC are overexpression of BCL-2 and inactivation of p53 and retinoblastoma (Rb) protein. Inactivation of p53 and Rb protein has been found to account up to 90% of SCLC cases, and the inactivation of the two proteins can be caused by a mutation (gene deletion).90 Other than differences in histological features between SCLC and NSCLC, these two subtypes of lung cancer also different in term of expression of neuroendocrine (NE) differentiation markers. NE differentiation markers only express in SCLC but not in NSCLC such as chromogranin A, neuron-specific enolase, synaptophysin, or neural cell adhesion molecule.91,92 Study on chemoprevention of SCLC is minimal, especially on chemopreventive effects of natural compounds toward SCLC. Difficulties in developing a mouse model that relevance to human SCLC can be due to the complexity of this type of lung cancers such as SCLC tumors contain different tumor type of cell populations (cells heterogeneity).93 Thus, the chemoprevention against SCLC was excluded from this review. Table 4 shows the summary chemotherapeutic researches of natural products against SCLC together with LCC.

Conclusion

Since numerous interventions have been done to improve the survival rate and burden of lung cancer patients, but lung cancer is still one of the major causes of cancer-related death. Other than focusing on improvement of treatment, chemoprevention of lung cancer has gained increasing popularity lately. Molecular targeted therapy specifically on each subtype of lung cancers might be one of the alternatives to improve the current treatments as it provides high efficacy approaches. Natural products are a great potential candidate for chemotherapeutic and chemoprevention as they have less harmful side effects and adverse reactions. Therefore, the discovery of new chemotherapeutic and chemopreventive agents are urgently needed to control lung cancer cases. This review of findings from in vitro and in vivo studies using human cell lines and experimental animal model specifically on each subtype of lung cancers may be a good source for further investigation, including the clinical study to discover the effective chemotherapeutic and chemoprevention agents for lung cancer.

Acknowledgements

We gratefully acknowledge funding support from National University of Malaysia (UKM) grant of GUP-2016-079. Special thanks to Norizah Awang from Institute of Medical Research (IMR) of Malaysia for her support. No potential conflict of interest was reported by the authors.

Conflict of Interest

There is no conflict of interest.

References

  1. The top ten causes of death. http://www.who.int/mediacentre/factsheets/fs310/en/. Accessed: Nov 2017.
  2. Weidepass E. Lifestyle and cancer risk. Journal of Preventive Medicine & Public Health. 2010;43(6):459-71.
    CrossRef
  3. Heikkilä K., Nyberg S. T., Theorell T., Fransson E. I., Alfredsson L., Bjorner J. B., et al. Work stress and risk of cancer: meta-analysis of 5700 incident cancer events in 116000 European men and women. British Medical Journal. 2013;346:165–175.
    CrossRef
  4. Field R. W., Withers B. L. Occupational and environmental causes of lung cancer. Clinics in Chest Medicine. 2012;33(4).
    CrossRef
  5. Pesch B., Kendzia B., Gustavsson P., Jockel K. H., Johnen G., Pohlabeln H., et al. Cigarette smoking and lung cancer-relative risk estimates for the major histological types from a pooled analysis of case-control studies. International Journal of Cancer. 2012;1;131(5):1210-9.
  6. Hoffmann D., Hoffmann I., El Bayoumy K. The less harmful cigarette: a controversial issue. Chemical Research in Toxicology. 2001;14:767–90.
    CrossRef
  7. Hecht S. S. Tobacco smoke carcinogens and lung cancer. Journal of the National Cancer Institute. 1999;91:1194–210.
    CrossRef
  8. Oser M. G., Neiderst M. J., Sequist L. V., Engelman J. A. Transformation from non-small cell lung cancer to small cell lung cancer: molecular drivers and cells of origin. Lancet Oncology. 2015;16(4):e165-72.
    CrossRef
  9. Campbell J. D., Alexandrov A., Kim J., Wala J., Berger A. H., Pedamallu C. S., et al. Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas. Nature Genetics. 2016;48(6):607–616.
    CrossRef
  10. Seve P., Reiman T., Dunmontet C. The role of βIII tubulin in predicting chemoresistance in non-small cell lung cancer. Lung Cancer. 2010;67:136-143.
    CrossRef
  11. Pan J., Zhang Q., Liu Q., Komas S. M., Kalyanaraman B., Lubet R. A., et al. Honokiol inhibits lung tumorigenesis through inhibition of mitochondrial function. Cancer Prevention Research. 2014;7(11):1149-59.
    CrossRef
  12. Chan C. S., Chan K. M. A review of lung cancer research in Malaysia. Medical Journal of Malaysia. 2016;71(1):70-78 9.
  13. Travis W. D. Classification of lung cancer. Semi Roentgenol. 2011;46(3):178-176.
    CrossRef
  14. Breuer R. H. J., Postmus P. E., Smit E. F. Molecular pathology of Non-small-cell lung cancer. Respiration. 2005;72:313-330.
    CrossRef
  15. Nesbitt J. C., Putnam Jr J. B., Walsh G. .L, Roth J. A., Mountain C. F. Survival in early-stage non-small cell lung cancer. The Annals of Thoracic Surgery. 1995;60:466-472.
    CrossRef
  16. Shah R., Sabanathan S., Richardson J., Meams A. J., Goulden C. Results of surgical treatment of stage I and II lung cancer. Journal of Cardiovascular Surgery (Torino). 1996;37(2):169-72.
  17. del Mar Delgado-Povedano M., Calderon-Santiago M., Priego-Capote F., Jurado-Gamez B., De Castro M. D. L. Recent advances in human sweat metabolomics for lung cancer screening. Metabolomics. 2016;12(11):166.
    CrossRef
  18. Walters S., Maringe C., Coleman M. P., Peake D., Butler J., Young N., et al. Lung cancer survival and stage at diagnosis in Australia, Canada, Denmark, Norway, Sweden and the UK: a population-based study, 2004-2007. Thorax. 2013;68(6):551-64.
    CrossRef
  19. Detterbeck F. C. The eight edition TNM stage classification for lung cancer: What does it mean on main street? Journal of Thoracic and Cardiovascular Surgery. 2018;155(1):357-359.
    CrossRef
  20. Zappa C., Mousa S. A. Non-small cell lung cancer: current treatment and future advances. Translational Lung Cancer Research. 2016;5(3):288-300.
    CrossRef
  21. Stewart L. A., Pignon J. P. Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials. British medical Journal. 1995;7;311(7010):899-909.
  22. Loanardo F., Li X., Kaplun A., Soubani A., Sethi S., Gadgeel S., et al. The natural tumor suppressor protein maspin and potential application in non-small cell lung cancer. Current Pharmaceutical Design. 2010;16(16):1877-1881.
    CrossRef
  23. Salomaa E. R., Sallinen S., Heikkanen H., Lippo K. Delays in the diagnosis and treatment of lung cancer. Chest. 2005;2282-2288.
    CrossRef
  24. Ramalingam S., Belani C. Systemic chemotherapy for advanced non-small cell lung cancer: recent advances and future directions. Oncologist. 2008;13(1):5-13.
    CrossRef
  25. Leighl N. B. Treatment paradigms for patients with metastatic non-small-cell lung cancer: first-, second- and third-line. Current Oncology. 2012;19(1):S52–S58.
    CrossRef
  26. Masters G. A., Temin S., Azzoli C. G., Giaccone G., Baker S. Jr., Brahmer J. R., et al. Systemic Therapy for Stage IV Non-Small-Cell Lung Cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. Journal of Clinical Oncology. 2015;20;33(30):3488-515.
  27. Yazbeck V. Y., Villaruz L., Haley M., Socinski M. A. Management of normal tissue associated with chemoradiation (primary skin esophagus, and lung). Cancer Journal. 2013;19(3):231-7.
    CrossRef
  28. Iwanmoto T. Clinical application of drug delivery systems in cancer chemotherapy: review of the efficacy and side effects of approved drugs. Biological and Pharmaceutical Bulletin. 2013;36(5) 715–718.
    CrossRef
  29. Walder D., O’Brien. Looking back and to the future: Are we improving ‘cure’ in non-small cell lung cancer? European Journal of Cancer. 2017;75:192-194.
    CrossRef
  30. Sporn M. B., Dunlop N. M., Newton D. L., Smith J. M. Prevention of chemical carcinogenesis by vitamin A and its synthetic analogs (retinoids). Federation Proceedings. 1976;1;35(6):1332-8.
  31. Siddiqui I. A., Sanna V., Ahmad N., Sechi M., Mukhtar H. Resveratrol nano formulation for cancer prevention and therapy. Annals of the New york Academy of Sciences. 2015;1348(1):20-31.
    CrossRef
  32. Gomperts B. N., Spira A., Massion P. P., Walser T. C., Wistuba I. I., Minna J. D., et al. Evolving concepts in lung carcinogenesis. Seminars in Respiratory and Critical Care Medicine. 2011;31(2): 32-43.
    CrossRef
  33. Dakubo G. D., Jakupciak J. P., Birch-Machin M. A., Parr R. L. Clinical implications and utility of field cancerization. Cancer Cell International. 2007;7:2.
    CrossRef
  34. Slaughter D. P., Southwick H. W., Smejkal W. Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer. 1953;6(5):963–968.
    CrossRef
  35. Zhang L., Lee J. J.,Tang H., Fan Y. H., Xioa L., Ren H., et al. Impact of smoking cessation on global gene expression in the bronchial epithelium of chronic smokers. Cancer Prevention Research. 2008;1(2):112–118.
    CrossRef
  36. Steiling K., Ryan J., Brody J. S., Spira A. The field of tissues injury in the lung and airway. Cancer Prevention Research. 2008;1(6):396-403.
    CrossRef
  37. Brandes J. C., Amin A. R. M. R., Khuri F., Shin D. M. Prevention of lung cancer: future perspective with natural compounds. Tuberculosis and Respiratory Diseases. 2010;69:1-15.
    CrossRef
  38. Tanveer S., Fathi E., Guy F. Towards new anticancer strategies by targeting cancer stem cells with phytochemical compounds. Cancer Stem Cells-The Cutting Edge.  2011;431–456.
  39. Bucher F. L., Bueno-de-Mesquita H. B., Linseisen J., Bosuizen H. C., Kiemeney L. A. L. M., Ros MM. Fruits and vegetables consumption and the risk of histological subtypes of lung cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC). Cancer Causes Control. 2010;21:357-371.
    CrossRef
  40. Lam T. K., Ruczinki I., Helzlsouer K. J., Shugart Y. Y., Caufield L. E., Alberg A. J. Cruciferous vegetable intake and lung cancer risk: A nested case-control study matched on cigarette smoking. Cancer Epidemiology, Biomarkers and Preventions. 2010;19(10):2534-40.
    CrossRef
  41. Garber M. E., Troyanskaya O. G., Schluens K., Petersen S., Thaesler Z., Pacyna-Gengelbach M., et al. Diversity of gene expression ion adenocarcinoma of the lung. Proceedings of National Academy of Science USA. 2001;98(24):13784-13789.
    CrossRef
  42. Tiseo M., Bartolotti M., Gelsomino F., Ardizzoni A. First-line treatment in advanced non-small-cell lung cancer: the emerging role of the histologic subtype. Expert Review of Anticancer Therapy. 2009;9:425–35.
    CrossRef
  43. Lortet-Tieulent J., Soerjomataram I., Ferlay J., Rutherford M., Weiderpass E., Bray F. International trends in lung cancer incidence by histological subtype: Adenocarcinoma stabilizing in men but still increasing in women. Lung Cancer. 2014;84(1):13-22.
    CrossRef
  44. Zhang B. C., Gao J., Wang J., Rao Z. G., Wang B. C., Gao J. F. Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogensis and poor prognosis in lung adenocarcinoma. Medical Oncology. 2011;28(4):1447-52.
    CrossRef
  45. Zhong L., Goldberg M. S., Gao Y. T & Jin F. A casecontrol study of lung cancer and environmental tobacco smoke among non-smoking women living in Shanghai. China. Cancer Causes Control. 1999;10(6):607-16.
    CrossRef
  46. Liam C. K., Lim K. H., Wong M. M. Lung cancer in patients younger than 40 years in multiracial Asian country. Respirology. 2000;5:355-361.
    CrossRef
  47. Herbst R. S., Heymach J. V., Scott M. L. Molecular origins of cancer: lung cancer. The New England. Journal of Medcine. 2008;359(13):1367-1380.
  48. Witsuba I. I. Molecular pathogenesis of non-small cell lung carcinomas. Journal of Lung Cancer. 2012;11(1):12-20.
    CrossRef
  49. Gazdar A. F., Shigematsu H., Herz J., Minna J. D. Mutations and addiction to EGFR: the Achille ‘heal’ of lung cancers? Trends in Molecular Medicine2012;10:481-486.
    CrossRef
  50. Le Calvez F., Mukeria A., Hunt J. D., Kelm O., Hung R. J., Taniere P., et al. TP53 and KRAS mutation load and types in lung cancers in relation to tabacco smoke: distinct patterns in never, former and current smokers. Cancer Research. 2005;15:65(12):5076-83.
  51. Paez J. G., Janne P. A., Lee J. C., Tracy S., Greulich H., Gabriel S., et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497-1500.
    CrossRef
  52. Roman M., Baraibar I., Lopez I., Nadal E., Rolfo C., Vicent S., et al. KRAS oncogene in non-small cell lung cancer: clinical perspective on the treatment of an old target. Molecular Cancer.2018;19;17(1):33.
  53. Chen R. J., Tsai S. J., Ho C. T., Pan M. H., Wu C. H., Wang Y. J. Chemopreventive effects of pterostilbene on urethane-induced lung carcinogenesis in mice via the inhibition of EGFR-mediated pathways and the induction of apoptosis and autophagy. Journal of Agriculture and Food Chemistry. 2012;60(46):11533-41.
    CrossRef
  54. Patolla J. M. R., Qian L., Biddick L., Zhang Y., Desai D., Amin S., et al. β-escin inhibits NNK-induced lung adenocarcinoma and ALDH1A1 and RhoA/Rock expression in A/J mice and growth of H460 human lung cancer cells. Cancer Prevention Research. 2013;6(10):1140-9.
    CrossRef
  55. Shukla S., Khan S., Kumar S., Sinha S., Farhan M., Bora H. K., et al. Cucurbitacin B alters the expression of tumor-related genes by epigenetic modification in NSCLC and inhibits NNK-induced lung tumorigenesis. Cancer Prevention Research. 2015;(6):552-62.
    CrossRef
  56. Alexandrow M. G., Song L. J., Altiok S., Gray J., Haura E. B., Kumar N. B. Curcumin: A novel Stat 3 pathway inhibitor for chemoprevention of lung cancer. European Journal of Cancer Prevention2012;21(5):407-412.
    CrossRef
  57. Singh T., Prasad R., Katiyar S. K. Inhibition of class I histone deacetylases in non-small cell lung cancer by honokiol leads to suppression of cancer cell growth and induction of cell death in vitro and in vivo. Epigenetics2013;1;8(1):54–65.
  58. He L., Bi J., Guo Q., Yu Y., Ye X. Effects of emodin extracted from Chinese Herbs on proliferation of non-small cell lung cancer and underlying mechanisms. Asian PacificJournal of Cancer Prevention. 2012;13:1505-1510.
    CrossRef
  59. Wei Y., Xu Y., Han X., Qi Y., Xu L., Xu Y., et al. Anti-cancer of dioscin on three kinds of human lung cancer cell lines through inducing DNA damage and activating mitochondrial signal pathway. Food and Chemical Toxicology. 2013;59(9):118-28.
    CrossRef
  60. Lin Y., Xu J., Liao H., Li L., Pan L. Piperine induces apoptosis of lung cancer A549 cells via p53-dependent mitochondrial signaling pathway. Tumour Biology. 2014;35(4):3305-10.
    CrossRef
  61. Li Y. C., He S. M., He Z. X., Li M., Yang Y., Pang J. X., et al. Plumbagin induces apoptotic and autophagic cell death through inhibition of the P13K/Akt/mTOR pathway in human non-small cell lung cancer cells. Cancer Letters. 2014;344(2):239-59.
    CrossRef
  62. Xue R., Han N., Xia M., Ye C., Hao Z., Wang L., et al. TX A9, cardiac glycoside Streptocaulon juventas, exerts a potent anti-tumor activity against human non-small ccell lung cancer cells in vitro and in vivo. Steroids. 2015;94(9):51-9.
    CrossRef
  63. Shi J., Liu F., Zhang W., Liu X., Lin B., Tang X. Epigallocatechin-3-gallate inhibits nicotine-induced migration and invasion by the suppression of angiogenesis and epithelial-mesenchymal transition in non-small cell lung cancer cells. Oncology Report. 2015;33(6):2972-80.
    CrossRef
  64. Zhang Q., Pan J., Lubet R. A., Wang Y & You M. Targeting the insulin-like growth factor-1 receptor by picropodophyllin for lung cancer chemoprevention. Molecular Carcinogonesis. 2015;54(1):129-37.
    CrossRef
  65. Perez-Moreno P., Brambilla E., Thomas R., Soria J. C. Squmaous cell carcinoma of the lung: molecular subtypes and therapeutic opportunities. Clinical Cancer Research. 2012;1;18(9):2443-51.
  66. Miller Y. E. Pathogenesis of lung cancer: 100 year report. American Journal of Respiratory Cell and  Molecular Biology. 2005;33(3):216-23.
    CrossRef
  67. Liam C. K., Pang Y. K., Leow C. H., Poosparajah S., Menon A. Changes in the distribution of lung cancer cell types and patient demography in developing multiracial Asian country: Experience of a university teaching hospital. Lung Cancer. 2006;53(1):23-30.
    CrossRef
  68. Khunder S. A. Effect of cigarette smoking on major histological types of lung cancer: a meta-analysis. Lung Cancer. 2001;31:139-148.
    CrossRef
  69. Kenfield S. A., Wei E. K., Stampfer M. J., Rosner B. A., Colditz G. A. Comparison of aspects of smoking among the four histological types of lung cancer. Tobacco Control. 2008;17(3):198-204.
    CrossRef
  70. Xu C., Fillmore C. M., Koyama S., Wu H., Zhao Y., Chen B., et al. Loss of Lkb1 and Pten leads to lung squamous cell carcinoma with elevated PD-L1 expression. Cancer Cell. 2014;12;25(5):590-604.
  71. Rekhtman N., Paik P. K., Arcila M. E., Tafe L. J., Oxnard G. R.,Moreira A. L., et al. Clarifying the spectrum of driver oncogene mutations in biomarker-verified squamous carcinoma of lung: lack of EGFR/KRAS and presence of PIK3CA/AKT1 mutations. Clinical Cancer Research. 2012;15;18(4):1167-76.
  72. Sandler A., Gray R., Perry M. C., Brahmer J., Schiller J. H., Dowlati A., et al. Paclitaxel-Carboplatin alone or with bevacizumb for non-small cell lung cancer. The New England Journal Medicine. 2006;335(24):2542-2550.
    CrossRef
  73. Pan J., Zhang Q., Liu Q., Komas S. M., Kalyanaraman B., Lubet R. A., et al. Honokiol inhibits lung tumorigenesis through inhibition of mitochondrial function. Cancer Prevention Research. 2014;7(11):1149-59.
    CrossRef
  74. Mazzilli S. A., Hershberger P. A., Reid M. E., Bogner P. N., Atwood K., Trump D. L., et al. Vitamin D repletion reduces the progression of premalignant squamous lesions in the NTCU lung squamous cell carcinoma mouse model. Cancer Prevention Research. 2015;8(10):895-904.
    CrossRef
  75. Zhao P., Pan Z., Luo Y., Zhang L., Li X., Zhang G., et al. Alantolactone induces apoptosis and cell cycle arrest on lung squamous cancer SK-MES-1 cells. Journal of Biochemistry and Molecular Toxicology. 2015;29(5):199-206.
    CrossRef
  76. Hua P., Zhang G., Zhang Y., Sun M., Cui R., Li X., et al. Custonolide induces G1/S phase arrest and activates mitochondrial-mediated apoptosis pathways in SK-MES 1 human lung squamous carcinoma cells. Oncology Letters. 2016;11:2780-2786.
    CrossRef
  77. Garber M. E., Troyanskaya O. G., Schluens K., Petersen S., Thaesler Z., Pacyna-Gengelbach M., et al. Diversity of gene expression in adenocarcinoma of the lung. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(24):13784-13789.
    CrossRef
  78. Rossi G., Mengoli M. C., Cavazza A., Nicoli D., Barbareshi M., Cantaloni C., et al. Large cell carcinoma of the lung: clinical oriented classification integrating immunohistochemistry and molecular biology. Virchows Archiv. 2014;464(1):61-8.
    CrossRef
  79. Gridelli C., Rossi A., Carbone D. P., Guarize J., Karachaliou N., Mok T., et al. Non-small cell lung cancer. Nature Reviews Disease Primer. 2015;1:15009.
    CrossRef
  80. Rekhtman N., Pietanza M. C., Hellmann M. D., Naidoo J., Arora A., Won H., et al. Next-generation sequencing of pulmonary large cell neuroendocrine carcinoma reveals small cell carcinoma-like and non-small cell carcinoma-like subsets. Clinical Cancer Research. 2016;15;22(14):3618-29.
  81. Rekhtman N. Neuroendocrine tumors of the lung: An update. Archives of Pathology & Laboratory Medicine. 2010;134(11):1628-38.
  82. Hung J. Y., Hsu Y. L., Ko Y. C., Tsai Y. M., Yang C. J., Huang M. S., et al. Didymin, a dietary flavonoid glycoside from citrus fruits, induces Fas-mediated apoptotic pathway in human non-small-cell lung cancer cells in vitro and in vivo. Lung Cancer. 2010;68(3):366-74.
    CrossRef
  83. Shieh J. M., Cheng T. H., Shi M. D., Wu P. F., Chen Y., Ko S. C., et al. α-tomatine suppresses invasion and migration of human non-small cell lung cancer NCI-H460 cells through inactivating FAK/PI3K/Akt signaling pathway and reducing binding activity of NF-кB. Cell Biochemistry and Biophysics. 2011;60(3):297-310.
    CrossRef
  84. Bi N., Cao J., Song Y., Shen J., Liu W., Fan J., et al. A microRNA signature predicts survival ion early stage small-cell lung cancer treated with surgery and adjuvant chemotheraphy. PloS One2014;9(3):91388.
    CrossRef
  85. Jackman D. M., Johnson B. E. Small-cell lung cancer. Lancet. 2005;366:1385-1396.
    CrossRef
  86. Fruh M., Ruyssher D. D., Popat S., Crino L., Peters S., Felip E. Small-cell lung cancer (SCLC): ESMO clinical practice guidelines for diagnosis, treatment and follow up. Annals of  Oncology. 2013;24(6):vi99-vi105.
    CrossRef
  87. Gazdar A. F., Bunn P. A., Minna J. D. Small-cell lung cancer: what we know, what we need to know and the path forward. Nature Reviews Cancer 2017;17(12):725.
    CrossRef
  88. Parsons A., Daley A., Begh R. Influence of smoking cessation after diagnosis of early stage lung cancer on prognostic: systemic review of observational studies with meta-analysis. British Medical Journal. 2010;340:5569.
    CrossRef
  89. Alexandrov L. B., Ju Y. S., Haase K., Loo P. V., Martincorena I., Nik-Zainal S., et al. Mutational signatures associated with tobacco smoking in human cancer. Science. 2016;354(6312):618-622.
    CrossRef
  90. Panani A., Roussos C. Cytogenetic and molecular aspects of lung cancer. Cancer Letters.  2006;239:1-9 9.
  91. Rom W. N., Hay J. G., Lee T. C., Jiang Y., Tchou-Wong K. M. Molecular and genetic aspects of lung cancer. American Journal of Respiratory and Critical Care Medicine. 2000;161:1355–1367.
    CrossRef
  92. Meuwissen R., Linn S. C., Linnoila R. I., Zevenhoven J., Mooi W. J., Berns A. Induction of small cell lung cancer by somatic inactivation of both Trp53 and Rb1 in a conditional mouse model. Cancer Cell. 2003;4:181-9.
    CrossRef
  93. Calbo J., van Montfrort E., Natalie P., van Drunen E., Beverloo H. B., Meuwissen R., et al. A functional role for tumor cell heterogeneity in mouse model of small cell lung cancer. Cancer Cell. 2011;19:244-256.
    CrossRef
  94. Yang C. L., Liu Y. Y., Ma Y. G., Xue Y. X., Liu D. G., Ren Y., et al. Curcumin blocks small cell lung cancer cells migration, invasion, angiogenesis, cell cycle and neoplasia through Janus Kinase-STAT3 signaling pathway. PLoS One. 2012;7(5):37960.
    CrossRef
  95. Zhong Z., Dong Z., Yang L., Chen X., Gong Z. Inhibition of proliferation of human lung cancer cells by green tea catechins is mediated by upregulation of let-7. Experimental and Therapeutic Medicine. 2012;4(2):267-272.
    CrossRef
  96. Lv C., Mioa L., Xu G., Wei S., Wang B., Huang C., et al. Wintilactone A as novel potential antitumor agent induces apoptosis and G2/M arrest of human lung carcinoma cells, and is mediated by HRas-GTP accumulation to excessively activate the Ras/Raf/ERK/p53-p21 pathway. Cell Death & Disease. 2013;5;4:952.
  97. Fang C., Zhang J., Qi D., Fan X., Luo J., Liu L., et al. Evodiamine induces G2/M arrest and apoptosis via mitochondrial and endoplasmic reticulum pathway in H446 and H1688 human small-cell lung cancer cells. PloS One. 2014;9(12):115204.
    CrossRef
Share Button
(Visited 1,573 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.