Abstract
Background:
The gradual evolution of the detection and quantification of volatile organic compounds (VOCs) has been instrumental in cancer diagnosis. The primary objective of this study was to assess the diagnostic potential of exhaled breath and urinary VOCs in cancer detection. As VOCs are indicative of tumor and human metabolism, our work also sought to investigate the metabolic pathways linked to the development of cancerous tumors.
Materials and Methods:
An electronic search was performed in the PubMed database. Original studies on VOCs within exhaled breath and urine for cancer detection with a control group were included. A meta-analysis was conducted using a bivariate model to assess the sensitivity and specificity of the VOCs for cancer detection. Fagan’s nomogram was designed to leverage the findings from our diagnostic analysis for the purpose of estimating the likelihood of cancer in patients. Ultimately, MetOrigin was employed to conduct an analysis of the metabolic pathways associated with VOCs in relation to both human and/or microbiota.
Results:
The pooled sensitivity, specificity and the area under the curve for cancer screening utilizing exhaled breath and urinary VOCs were determined to be 0.89, 0.88, and 0.95, respectively. A pretest probability of 51% can be considered as the threshold for diagnosing cancers with VOCs. As the estimated pretest probability of cancer exceeds 51%, it becomes more appropriate to emphasize the ‘ruling in’ approach. Conversely, when the estimated pretest probability of cancer falls below 51%, it is more suitable to emphasize the ‘ruling out’ approach. A total of 14, 14, 6, and 7 microbiota-related VOCs were identified in relation to lung, colorectal, breast, and liver cancers, respectively. The enrichment analysis of volatile metabolites revealed a significant enrichment of butanoate metabolism in the aforementioned tumor types.
Conclusions:
The analysis of exhaled breath and urinary VOCs showed promise for cancer screening. In addition, the enrichment analysis of volatile metabolites revealed a significant enrichment of butanoate metabolism in four tumor types, namely lung, colorectum, breast and liver. These findings hold significant implications for the prospective clinical application of multiomics correlation in disease management and the exploration of potential therapeutic targets.
Keywords: cancer, diagnosis, electronic nose, microbiota, pathway analysis, volatile organic compounds
Introduction
Highlights
The pooled sensitivity, specificity, and the area under the curve for cancer screening utilizing exhaled breath and urine volatile organic compounds (VOCs) were determined to be 0.89, 0.88, and 0.95, respectively.
A pretest probability of 51% can be considered as the threshold for diagnosing cancers with VOCs. As the estimated pretest probability of cancer exceeds 51%, it becomes more appropriate to emphasize the ‘ruling in’ approach. Conversely, when the estimated pretest probability of cancer falls below 51%, it is more suitable to emphasize the ‘ruling out’ approach.
A total of 14, 14, 6, and 7 microbiota-related VOCs were identified in relation to lung, colorectal, breast, and liver cancers, respectively.
The enrichment analysis of volatile metabolites revealed a significant enrichment of butanoate metabolism in four tumor types, namely lung, colorectum, breast, and liver.
Cancer poses a global public health concern and remains the leading cause of mortality worldwide1. The identification of cancer at an early stage, coupled with effective treatment, has the potential to significantly enhance survival rates among patients2. Metabolomics has emerged as a valuable methodology for characterizing the metabolic profiles of diseases, aiming to identify biomarkers for disease screening and early detection3. In order to address these challenges, the field of volatolomics, which involves the analysis of volatile organic compounds (VOCs) originating from both host and microbial metabolism and detectable in diverse biological matrices, presents a promising advancement in cancer biomarker-focused strategies4–6.
Several methodologies have been employed in the examination of VOCs in order to ascertain cancer biomarkers. Gas chromatography coupled with mass spectrometry (GC-MS) has been extensively utilized for the detection of VOCs associated with cancer7. Although GC-MS is informative, its analysis procedure is time-consuming and necessitates highly skilled personnel. Electronic nose (e-Nose) is another analytical method that can extract complete information from exhaled breath or urine components instead of focusing on the identification of specific biomarkers. The fundamental component of the e-Nose apparatus, the sensor array, can be constructed using a range of sensors, such as nanomaterials, metal oxide semiconductor (MOS) sensors, and other specialized sensors capable of detecting specific VOCs8,9. The analysis of exhaled breath using e-Nose technologies has been the subject of extensive research in the field of oncology. However, it is important to note that the accuracy of e-Nose results can be significantly affected by both endogenous and exogenous factors. Therefore, further exploration is required to enhance its precision10.
VOCs present in the human volatilome originate from both endogenous biochemical processes and environmental exposures, and are released through various biological matrices, including exhaled breath and urine11. Breath, being the most prevalent source of VOCs, has been subject to extensive scrutiny regarding its potential association with lung cancer12. Numerous studies in the literature have demonstrated that the presence of VOCs in breath may serve as a reflection of the biochemical condition of the human body, thereby enabling disease diagnosis. In addition to breath, more than 10% of VOCs associated with humans and tumors can be detected in urine11. The collection of urine presents advantages over breath collection and storage, and urine can be conveniently stored frozen for extended periods, facilitating the tracking of subjects. These characteristics render urine an optimal sample for the investigation of VOC profiling13.
The objective of this present study was to conduct a comprehensive review and meta-analysis of prior research on VOCs found in exhaled breath and urine, with the purpose of detecting cancer. As VOCs are indicative of tumor and human metabolism, our work also sought to investigate the metabolic pathways linked to the development of cancerous tumors.
Materials and methods
This study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement (Supplemental Digital Content 1, http://links.lww.com/JS9/B586) (Supplemental Digital Content 2, http://links.lww.com/JS9/B587) and AMSTAR-2 guidelines14,15 (Supplemental Digital Content 3, http://links.lww.com/JS9/B588).
Literature search strategy
We conducted a comprehensive literature search using the PubMed database to determine the overall diagnostic performance of VOCs in detecting tumors. The following search terms were used: (volatile organic compounds OR VOCs) AND [Neoplasms (Mesh) OR cancer OR carcinoma OR tumor OR neoplasm]. The search included studies published up until 30 June 2023, without any lower date limit.
Eligibility and exclusion criteria
The inclusion criteria for diagnostic studies were as follows: (1) utilization of either e-Nose technology or mass spectrometry; (2) cancer screening; and (3) examination of exhaled breath or urine. Conversely, studies were excluded if they met any of the following criteria: (1) not conducted on human subjects; (2) analysis of alternative biofluids such as blood or feces; and (3) inadequate provision of information required for diagnostic value calculations.
Data extraction and quality assessment
The data extraction process involved the participation of two reviewers (Q.W. and X.L.), with one initially extracting the data and the other conducting a double-check. The following data of each study were collected: author, nation, publication year, cancer type and stage, control group, sample size, analysis methodology (exhaled breath or urine, analytical platform and statistical method), results of true positive (Tp), false positive (Fp), false negative (Fn), and true negative (Tn) (either found or calculated from data in original published studies), and identified unique VOCs (if provided). The evaluation of study quality was conducted by two researchers who utilized the QUality Assessment of Diagnostic Accuracy Studies (QUADAS-2) checklist tool. Studies with a total score of nine points or higher were selected, and any discrepancies were resolved through consensus.
Pathway analysis
MetOrigin is an all-encompassing platform specifically designed for discerning the origins of metabolites by means of an integrated database search16. Subsequently, a compilation of metabolites exhibiting significant alterations, along with their corresponding Kyoto Encyclopedia of Genes and Genomes and Human Metabolome Database identification numbers, was uploaded, leading to the execution of the Simple MetOrigin Analysis.
Statistical analysis
Diagnostic parameters for VOCs were computed using the following formulas in each study: sensitivity=Tp divided by the sum of Tp+Fn, specificity=Tn divided by the sum of Tn+Fp, positive likelihood ratio (PLR)=sensitivity divided by one minus specificity (1-specificity), negative likelihood ratio (NLR)=one minus sensitivity (1-sensitivity) divided by specificity, and diagnostic odds ratio=Tp multiplied by Tn divided by Fn multiplied by Fp. Additionally, their 95% CI were determined. The bivariate model was adjusted to calculate the area under the curve (AUC). Fagan’s nomogram was designed to leverage the findings from our diagnostic analysis for the purpose of estimating the likelihood of cancer in patients. In order to identify the potential determinants of accuracy estimates, we endeavored to investigate the underlying causes of variability among the studies included in our analysis, specifically focusing on those with a quantified I 2 value exceeding 50%. Given the significance of the threshold effect as a contributing factor to heterogeneity, we conducted an assessment using Spearman’s correlation coefficient, whereby a negative correlation (P<0.05) would indicate the presence of said threshold effect. In the absence of a threshold effect but with the presence of significant heterogeneity, additional meta-regression analysis and subgroup analysis were conducted to investigate alternative factors contributing to heterogeneity in the studies included17. Additionally, Deeks funnel plot symmetry test was employed to directly assess publication bias18. The statistical analyses were performed using Stata 17.0 (StataCorp), with all tests being two-sided and a P-value less than 0.05 considered as statistically significant.
Results
Study characteristics
The literature search resulted in the identification of 85 publications that satisfied the predetermined inclusion criteria (Table 1)19–103. Out of these, five studies provided data for 10 separate comparisons, resulting in the incorporation of a cumulative total of 90 studies (with a combined number of cases in tumor and control groups amounting to 6077 and 8004, respectively) in our research. The process of study selection was visually depicted in Figure 1 through a detailed flowchart. The included studies examined various types of cancer, including lung cancer (n=38), colorectal cancer (n=11), urologic neoplasms (n=9 (prostate cancer=4, bladder cancer=3, renal cancer=2)), breast cancer (n=8), gastroesophageal cancer (n=7), head and neck cancer [n=7 (head and neck squamous cell carcinoma=5, thyroid cancer=2)], hepatocellular carcinoma (n=4), pancreatic cancer (n=2), ovarian cancer (n=1), cervical cancer (n=1), malignant pleural mesothelioma (n=1), and malignant mesothelioma (n=1). The majority of the included studies conducted comparisons between patients diagnosed with cancer, often exhibiting a combination of histologic subtypes, and either a healthy control group or patients with benign conditions that impacted the same organ. These studies typically encompassed patients at both early and advanced tumor stages, although 34 studies did not provide information on tumor stage. Frequently employed analytical platforms in this study encompassed e-Nose (n=41) and GC-MS (n=25). Among the e-Noses, the Cyranose 320 (n=8) and Aeonose (n=6) were the most prevalent. Additionally, a diverse assortment of custom-made e-Noses were utilized. The predominant sensor type employed was MOS (n=15). MOS was utilized in Aeonose, PEN3, and various e-nose prototypes for the detection of VOCs. Other reported sensor types included nanomaterial-based sensors (n=8) and quartz microbalance sensor (n=1).
Table 1.
Characteristics and outcomes of all 85 publications included in the qualitative analysis.
| Study ID | Nation | Tumor type | Tumor stage | Control group | No. of patients | Test sample | Analytical platform | Statistical method | Tp | Fp | Fn | Tn |
|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Phillips et al. 199919 | USA | Lung cancer | I | Cancer-free controls | 9, 48 | Exhaled breath | GC-MS | FSDA | 9 | 9 | 0 | 39 |
| Phillips et al. 2003a20 | USA | Lung cancer | NR | Healthy controls | 67, 41 | Exhaled breath | GC-MS | FSDA | 60 | 7 | 7 | 34 |
| Phillips et al. 2003b21 | USA | Breast cancer | NR | Healthy controls | 51, 42 | Exhaled breath | GC-MS | FSDA | 48a | 11a | 3a | 31a |
| Machado et al. 200522 | USA | Lung cancer | I–IV | Cancer-free controls | 14, 62 | Exhaled breath | e-Nose (Cyranose 320: 32 conducting polymer sensors) | SVM | 10 | 5 | 4 | 57 |
| Phillips et al. 200823 | USA | Lung cancer | NR | Cancer-free controls | 193, 211 | Exhaled breath | GC-MS | WDA | 163 | 40 | 30 | 171 |
| Ligor et al. 200924 | Austria | Lung cancer | NR | Healthy controls | 65, 31 | Exhaled breath | GC-MS | NR | 33 | 0 | 32 | 31 |
| Song et al. 201025 | China | Lung cancer | NR | Healthy controls | 43, 41 | Exhaled breath | GC-MS | NR | 41 | 6 | 2 | 35 |
| Qin et al. 201026 | China | Hepatocellular carcinoma | I–IV | Healthy controls | 30, 36 | Exhaled breath | GC-MS | LDA | 26 | 3 | 4 | 33 |
| Phillips et al. 201027 | USA | Breast cancer | NR | Healthy controls | 14, 69 | Exhaled breath | GC-MS | WDA | 12a | 14a | 2a | 55a |
| Dragonieri et al. 201228 | Italy | Malignant pleural mesothelioma | I–IV | Asbestos exposure | 13, 13 | Exhaled breath | e-Nose (Cyranose 320: 32 conducting polymer sensors) | CDA | 12 | 2 | 1 | 11 |
| Peled et al. 201229 | Israel | Lung cancer | I–IV | Benign cases | 50, 19 | Exhaled breath | e-Nose [18 nanomaterial sensors (GNPs and SWCNTs)] | DFA | 43 | 1 | 7 | 18 |
| Chapman et al. 201230 | Australia | Malignant mesothelioma | Advanced stage | Healthy controls | 10, 32 | Exhaled breath | e-Nose (Cyranose 320: 32 conducting polymer sensors) | PCA | 9b | 3b | 1b | 29b |
| Altomare et al. 201331 | Italy | Colorectal cancer | I–IV | Healthy controls | 37, 41 | Exhaled breath | GC-MS | PNN | 32b | 7b | 5b | 34b |
| Broza et al. 201332 | Israel | Lung cancer | Early stage | Benign cases | 12, 5 | Exhaled breath | e-Nose (25 nanomaterial sensors: GNP and PtNPs) | DFA | 12 | 1 | 0 | 4 |
| Xu et al. 201333 | China | Gastric cancer | I–IV | Benign gastric ulcers and less severe gastric conditions | 37, 93 | Exhaled breath | e-Nose [14 nanomaterial sensors (GNP and SWCNTs)] | DFA | 33 | 9 | 4 | 84 |
| Handa et al. 201434 | Japan | Lung cancer | I–IV | Healthy controls | 50, 39 | Exhaled breath | IMS | NR | 38 | 0 | 12 | 39 |
| Arasaradnam et al. 201435 | UK | Colorectal cancer | I–IV | Healthy controls | 83, 50 | Urine | FAIMS | FDA | 73 | 20 | 10 | 30 |
| Bousamra et al. 201436 | USA | Lung cancer | 0–II | Benign pulmonary diseases | 64, 40 | Exhaled breath | FT-ICR-MS | NR | 53 | 10 | 11 | 30 |
| Gruber et al. 201437 | Israel | HNSCC | I–IV | Healthy controls | 22, 19 | Exhaled breath | e-Nose (6 nanomaterial sensors) | DFA | 17 | 2 | 5 | 17 |
| Fu et al. 201438 | China | Lung cancer | I–IV | Benign pulmonary nodules | 97, 32 | Exhaled breath | FT-ICR-MS | NR | 87 | 6 | 10 | 26 |
| Leunis et al. 201439 | The Netherlands | HNSCC | I–IV | Benign cases | 36, 23 | Exhaled breath | e-Nose (12 MOS) | LRA | 32 | 5 | 4 | 18 |
| Schumer et al. 201540 | USA | Lung cancer | NR | Healthy controls | 156, 194 | Exhaled breath | silicon chip-MS | NR | 146 | 28 | 10 | 166 |
| Li et al. 201541 | China | Lung cancer | I–IV | Benign pulmonary nodules and healthy controls | 85, 119 | Exhaled breath | FT-ICR-MS and GC-MS | PLS, SVM, RF, LDA, and QDA | 82 | 19 | 3 | 100 |
| Ligor et al. 201542 | Poland | Lung cancer | III–IV | Healthy controls | 123, 361 | Exhaled breath | GC-MS | ANN | 78b | 100b | 45b | 261b |
| Kumar et al. 201543 | UK | Esophageal and gastric adenocarcinoma | I–IV | Healthy controls | 81, 129 | Exhaled breath | SIFT-MS | LRA | 71 | 24 | 10 | 105 |
| Amal et al. 201544 | Israel | Ovarian cancer | I–IV | Cancer-free controls | 34, 34 | Exhaled breath | GC-MS | DFA | 30b | 3b | 4b | 31b |
| Guo et al. 201545 | China | Papillary thyroid carcinoma | NR | Nodular goiters | 25, 39 | Exhaled breath | GC-MS | PCA, PCA, and PLS-DA | 23 | 7 | 2 | 32 |
| Barash et al. 201546 | Israel | Breast cancer | NR | Benign cases and healthy controls | 169, 82 | Exhaled breath | e-Nose [40 nanomaterial sensors (GNP and SWCNTs)] | DFA | 132 | 32 | 37 | 50 |
| Rocco et al. 201647 | Italy | Lung cancer | Advanced stage | Healthy controls | 23, 77 | Exhaled breath | e-Nose (BIONOTE: 7 acoustic-mass sensors) | PLS-DA | 20 | 4 | 3 | 73 |
| Amal et al. 2016a48 | Israel | Colorectal cancer | I–IV | Healthy controls | 65, 122 | Exhaled breath | e-Nose [6 nanomaterial sensors (GNP and SWCNTs)] | DFA | 55 | 7 | 10 | 115 |
| Amal et al. 2016b49 | Israel | Gastric cancer | I–IV | Gastric intestinal metaplasia | 30, 95 | Exhaled breath | e-Nose [8 nanomaterial sensors (GNP and SWCNTs)] | DFA | 22a | 2a | 8a | 93a |
| Schallschmidt et al. 201650 | Germany | Lung cancer | NR | Healthy controls | 37, 23 | Exhaled breath | GC-MS | LDA | 34 | 1 | 3 | 22 |
| Zou et al. 201651 | China | Esophageal cancer | I–IV | Healthy controls | 29, 57 | Exhaled breath | PTR-MS | SDA | 25 | 6 | 4 | 51 |
| Gasparri et al. 201652 | Italy | Lung cancer | NR | Healthy controls | 70, 76 | Exhaled breath | e-Nose (8 QMB sensors) | PLS-DA | 57 | 7 | 13 | 69 |
| Sakumura et al. 201753 | Japan | Lung cancer | I–IV | Healthy controls | 107, 29 | Exhaled breath | GC-MS | SVM | 102 | 3 | 5 | 26 |
| Li et al. 201754 | China | Lung cancer | NR | Healthy controls and benign cases | 24, 28 | Exhaled breath | e-Nose (14 sensors, MOS HWG, CCGS, EGS) | LDA (fuzzy 5-NN) | 22 | 2 | 2 | 26 |
| Shlomi et al. 201755 | Israel | Lung cancer | Early stage | Benign nodules | 16, 30 | Exhaled breath | e-Nose (40 nanomaterial sensors: 26 GNP, 8 SWCNT/PAH, and 6 SWCNT/HBC) | DFA | 12 | 2 | 4 | 28 |
| Schuermans et al. 201856 | China | Gastric cancer | NR | Healthy controls | 16, 28 | Exhaled breath | e-Nose (3 MOS) | ANN | 13 | 8 | 3 | 20 |
| Heers et al. 201857 | Germany | Bladder transitional cell carcinoma | I–IV | Healthy controls | 30, 30 | Urine | e-Nose (Cyranose 320: 32 composite polymer sensors) | LDA | 28 | 2 | 2 | 28 |
| Markar et al. 201858 | UK | Pancreatic cancer | NR | Cancer-free controls | 25, 43 | Exhaled breath | GC-MS | LRA | 20b | 2b | 5b | 41b |
| Widlak et al. 201859 | UK | Colorectal cancer | NR | Cancer-free controls | 35, 406 | Urine | FAIMS | PCA | 22 | 151 | 13 | 255 |
| Marzorati et al. 201960 | Italy | Lung cancer | Early stage | Healthy controls | 6, 10 | Exhaled breath | e-Nose (4 MOS) | ANN (LOOCV) | 5 | 0 | 1 | 10 |
| Nissinen et al. 201961 | Finland | Pancreatic cancer | I–IV | Healthy controls | 68, 52 | Urine | FAIMS | LDA | 54 | 11 | 14 | 41 |
| Rudnicka et al. 201962 | Poland | Lung cancer | I–IV | Healthy controls | 90, 91 | Exhaled breath | GC-MS | DFA | 72a | 8a | 18a | 83a |
| Chandran et al. 201963 | Australia | HNSCC | NR | Healthy controls | 23, 21 | Exhaled breath | SIFT-MS | NR | 21 | 5 | 2 | 16 |
| Mozdiak et al. 201964 | UK | Colorectal cancer | NR | Healthy controls | 12, 12 | Urine | FAIMS | NR | 12 | 1 | 0 | 11 |
| Mohamed et al. 201965 | Egypt | Lung cancer | I–IV | Without lung lesions | 28, 20; 24, 27c | Exhaled breath; urine | e-Nose (PEN3: 10 MOS) | PCA and ANN | 26; 24 | 2; 0 | 2; 0 | 18; 27 |
| Diaz de Leon Martinez et al. 202066 | Mexico | Breast cancer | 0-IV | Healthy controls | 262, 181 | Exhaled breath | e-Nose (Cyranose 320: 32 conducting polymer sensors) | CDA | 262 | 0 | 0 | 181 |
| Chen et al. 202067 | China | Lung cancer | NR | Healthy controls | 24, 25 | Exhaled breath | e-Nose (GO sensor) | PCA | 23 | 1 | 1 | 24 |
| van Keulen et al. 202068 | The Netherlands | Colorectal cancer | I–IV | Normal colonoscopy cases | 62, 104 | Exhaled breath | e-Nose (Aeonose: 3 MOS) | ANN (Aethena) | 59 | 37 | 3 | 67 |
| Waltman et al. 202069 | The Netherlands | Prostate cancer | I–IV | Healthy controls | 32, 53 | Exhaled breath | e-Nose (Aeonose: 3 MOS) | ANN | 27 | 16 | 5 | 37 |
| Kort 2020a70 | The Netherlands | NSCLC | NR | Healthy controls | 103, 84 | Exhaled breath | e-Nose (Aeonose: 3 MOS) | ANN | 95 | 41 | 8 | 43 |
| Altomare et al. 202071 | Italy | Colorectal cancer | I–IV, unknown | Cancer-free controls | 82, 87 | Exhaled breath | GC-MS | LRA | 74 | 6 | 8 | 81 |
| Gharra et al. 202072 | China | Lung and gastric cancers | I–IV | Healthy controls | 156, 106; 117, 109d | Exhaled breath | e-Nose (15 nanomaterial-based sensors) | DFA | 156b e; 117b e | 3b e; 2b e | 0b e; 0b e | 103b e; 107b e |
| Zhu et al. 202073 | UK | Bladder cancer | Ta, T1-2 | Healthy controls | 38, 41 | Urine | e-Nose (fluorescence sensitive optical array) | PLS-DA | 32 | 5 | 6 | 36 |
| Kort 2020b74 | The Netherlands | Lung cancer | I–IV | Suspected lung cancer and healthy controls | 138, 143 | Exhaled breath | e-Nose (3 MOS) | ANN | 130 | 80 | 8 | 63 |
| Tyagi et al. 2021a75 | UK | Prostate and bladder cancers | NR | Cancer-free controls | 15, 36; 55, 36f | Urine | GC-IMS | XGBoost | 13; 42 | 3; 4 | 2; 13 | 33; 32 |
| Capelli et al. 202176 | Italy | Prostate cancer | NR | Healthy controls | 132, 60 | Urine | e-Nose (6 MOS) | RF | 108 | 8 | 24 | 52 |
| Tyagi et al. 2021c77 | UK | Colorectal cancer | NR | Cancer-free controls | 58, 38 | Urine | e-Nose (10 MOS); GC-TOF-MS | ANN | 53g; 50g | 17g; 7g | 5g; 8g | 21g; 31g |
| Yang et al. 202178 | Taiwan, China | Breast cancer | NR | Cancer-free controls | 351, 88 | Exhaled breath | e-Nose (Cyranose 320: 32 conducting polymer sensors) | RF | 301 | 3 | 50 | 85 |
| Hong et al. 202179 | China | Gastric cancer | NR | Healthy controls | 29, 24 | Exhaled breath | TD-SPI-MS | LRA | 28 | 1 | 1 | 23 |
| V A 2021a80 | India | Lung cancer | NR | Healthy controls | 41, 74 | Exhaled breath | e-Nose (1 MOS) | XGBoost | 38b | 5b | 3b | 69b |
| Diaz de Leon Martinez et al. 202181 | Mexico | Cervical cancer | NR | Healthy controls | 12, 12 | Urine | GC-MS | CAP | 11 | 0 | 1 | 12 |
| V A 2021b82 | India | Lung cancer | I–III | Healthy controls | 32, 72 | Exhaled breath | e-Nose (5 MOS) | SVM | 30a | 3a | 2a | 69a |
| Bannaga et al. 202183 | UK | Hepatocellular carcinoma | NR | Nonfibrotic cases | 20, 31 | Urine | GC-IMS and GC-TOF-MS | RF | 12 | 8 | 8 | 23 |
| Pinto et al. 202184 | Portugal | Clear cell renal cell carcinoma | I–IV | Cancer-free controls | 75, 75 | Urine | GC-MS | PCA and PLS-DA | 62 | 16 | 13 | 59 |
| Long et al. 202185 | China | Lung cancer | NR | Healthy controls | 161, 116 | Exhaled breath | GC-MS | OPLS-DA | 149b e | 6b e | 12b e | 110b e |
| Chen et al. 202186 | China | Lung cancer | I–IV, unknown | Healthy controls | 101, 134 | Exhaled breath | e-Nose (11 gas sensors and 2 temperature and humidity sensors) | KPCA-XGBoost | 97e | 12e | 4e | 122e |
| Mohamed et al. 202187 | Sudan | HNSCC | I–IV | Healthy controls | 49, 35 | Exhaled breath | e-Nose (Aeonose: 3 MOS) | ANN (Aethena) | 43 | 10 | 6 | 25 |
| Boulind et al. 202288 | UK | Colorectal cancer | NR | Cancer-free controls | 18, 65 | Urine | GC-MS | ANN | 15 | 12 | 3 | 53 |
| Cheng et al. 202289 | The Netherlands | Colorectal cancer | I–IV | Healthy controls | 30, 84 | Exhaled breath | TD-SPI-MS | PCoA | 24 | 11 | 6 | 59 |
| Gasparri et al. 202290 | Italy | Lung cancer | I–III | Healthy controls | 46, 81 | Urine | GC-IMS; e-Nose (12 QMB sensors) | SVM | 46b e h; 43b e h | 3b e h; 1b e h | 0b e h; 3b e h | 78b e h; 80b e h |
| Scheepers et al. 202291 | The Netherlands | Differentiated thyroid carcinoma | NR | Benign cases | 48, 85 | Exhaled breath | e-Nose (Aeonose: 3 MOS) | NR | 35 | 26 | 13 | 59 |
| Sukaram et al. 202292 | Thailand | Hepatocellular carcinoma | A–D | Healthy controls and cirrhosis | 61, 91 | Exhaled breath | GC-MS | SVM | 46b | 16b | 15b | 75b |
| Gashimova et al. 202293 | Russia | Lung cancer | NR | Young healthy controls | 77, 78 | Exhaled breath | GC−MS | ANN | 68b e | 13b e | 9b e | 65b e |
| Anzivino et al. 202294 | Italy | HNSCC | NR | Allergic rhinitis case and healthy controls | 15, 30 | Exhaled breath | e-Nose (Cyranose 320: 32 polymer sensors) | PCA and CDA | 14 | 4 | 1 | 26 |
| Liu et al. 202395 | China | Prostate cancer | NR | Cancer-free controls | 23, 23 | Urine | GC-IMS | SVM | 20a | 1a | 3a | 22a |
| Costantini et al. 202396 | Italy | Renal cancer | I–III | Healthy controls | 110, 142 | Urine | e-Nose (Cyranose 320: 32 organic polymer sensors) | PCA | 79 | 15 | 31 | 127 |
| Li et al. 202397 | China | Breast cancer | NR | healthy controls | 25, 26 | Urine | HS-SPME and GC-HRMS | OPLS-DA | 19a | 2a | 6a | 24a |
| Nakayama et al. 202398 | Japan | Breast cancer | I–IV | Cancer-free controls | 45, 51 | Exhaled breath | SIFT-MS | MLR | 39e | 22e | 6e | 29e |
| Liu et al. 202399 | China | Breast cancer | I–IV, unknown | Healthy controls | 249, 1545 | Exhaled breath | HPPI-TOFMS | RF | 222a | 190a | 27a | 1355a |
| Temerdashev et al. 2023100 | Russia | Lung cancer | I–IV | Healthy controls | 78, 84 | Exhaled breath | GC-MS | ANN | 69e | 13e | 9e | 71e |
| Sani et al. 2023101 | China | Lung cancer | Early stage | Healthy controls | 291, 95 | Exhaled breath | LC-MS/MS | RF | 241 | 21 | 50 | 74 |
| Sukaram et al. 2023102 | Thailand | Hepatocellular carcinoma | A–C | Healthy controls and cirrhosis | 124, 219 | Exhaled breath | GC-FAIMS | XGBoost | 87 | 25 | 37 | 194 |
| Kort et al. 2023103 | The Netherlands | NSCLC | I–IV | Healthy controls | 158, 216 | Exhaled breath | e-Nose (Aeonose: 3 MOS) | ANN, SVM, RF, XGBoost, and logistic regression | 147b | 99b | 11b | 117b |
Data derived from a validated model.
Data derived from a training model.
Exhaled breath: lung cancer n=28, without lung lesions n=20; urine: lung cancer n=24, without lung lesions n=27.
Lung cancer n=156, healthy controls n=106; gastric cancer n=117, healthy controls n=109.
Diagnostic values were estimated based on calculable data in the study.
Prostate cancer n=15, cancer-free controls n=36; bladder cancer n=55, cancer-free controls n=36.
eNose: Tp, Fp, Fn, Tn n=53, 17, 5, 21; GC-TOF-MS: Tp, Fp, Fn, Tn n=50, 7, 8, 31.
GC-IMS: Tp, Fp, Fn, Tn n=46, 3, 0, 78; e-Nose: Tp, Fp, Fn, Tn n=43, 1, 3, 80.
ANN, artificial neural network; CAP, canonical principal coordinate analysis; CDA, canonical discriminant analysis; DFA, discriminant factor analysis; FAIMS, field asymmetric ion mobility spectrometry; FDA, fisher discriminant analysis; Fn, false negative; Fp, false positive; FSDA, forward stepwise discriminant analysis; FT-ICR-MS, fourier transform-ion cyclotron resonance-mass spectrometry; GC-FAIMS, gas chromatography-field asymmetric ion mobility spectrometry; GC-HRMS, gas chromatography-high resolution mass spectrometry; GC-MS, gas chromatography-mass spectrometer; GC-TOF-MS, gas chromatography-time of flight-mass spectrometry; HNSCC, head and neck squamous cell carcinoma; HPPI-TOFMS, high-pressure photon ionization-time-of-fight mass spectrometry; HS-SPME, headspace-solid phase microextraction; IMS, ion mobility spectrometry; KPCA, kernel principal component analysis; LDA, linear discriminant analysis; LRA, logistic regression analysis; MLR, multiple logistic regression; MOS, metal oxide semiconductor; NR, not reported; NSCLC, nonsmall cell lung cancer; OPLS-DA, orthogonal partial least-squares-discriminant analysis; PCA, principal component analysis; PCoA, principal co-ordinates analysis; PLS, partial least-squares; PLS-DA, partial least-squares-discriminant analysis; PNN, probabilistic neural network; PTR-MS, proton transfer reaction-mass spectrometry; QDA, quadratic discriminant analysis; QMB, quartz microbalance; RF, random forest; SDA, stepwise discriminant analysis; SIFT-MS, selected ion flow tube mass spectrometry; SVM, support vector machine; TD-SPI-MS, single-photon ionization-mass spectrometry coupled with on-line thermal desorption; Tn, true negative; Tp, true positive; WDA, weighted digital analysis; XGBoost, eXtreme Gradient Boosting.
Figure 1.
Flow diagram showing the study selection process.
Assessment of study quality and publication bias
The evaluation results of each publication based on QUADAS-2 were presented in Supplementary Table S1 (Supplemental Digital Content 4, http://links.lww.com/JS9/B589). The Deeks test for funnel plot symmetry revealed the absence of any significant publication bias (bias=0.73, P=0.469). Figure 2 displayed the corresponding Deeks plot, also known as funnel plot.
Figure 2.
Asymmetrical funnel plots indicated no publication bias (bias=0.73, P=0.469).
Diagnostic performance
A comprehensive analysis of receiver operating characteristic data from 90 studies revealed a pooled sensitivity of 0.89 (95% CI: 0.87–0.91) and specificity of 0.88 (95% CI: 0.85–0.90) (Figs 3–4). The pooled studies exhibited substantial heterogeneity, as indicated by an I 2 index of 84.70% for sensitivity and 92.51% for specificity (Fig. 3). Notably, there was no evidence of a threshold effect (Spearman correlation coefficient=0.32, P=0.10). The study findings revealed that the PLR, NLR, and AUC had values of 7.4 (95% CI: 6.0–9.1), 0.13 (95% CI: 0.10–0.15), and 0.95 (95% CI: 0.92–0.96), respectively. Additionally, Table 2 presented the diagnostic parameters for various types of cancers, including lung cancer, colorectal cancer, urologic neoplasms, breast cancer, gastroesophageal cancer, head and neck cancer, and hepatocellular carcinoma. The pretest probability was determined to be 0.25, 0.51, and 0.75, resulting in corresponding positive predictive values and negative predictive values of 71, 88, 96, and 4, 12, 28%, respectively (Fig. 5).
Figure 3.
Pooled sensitivity and specificity analyses of all 90 studies.
Figure 4.
Summary receiver operating characteristic curve analysis of all studies, the pooled area under the curve was 0.95 (95% CI: 0.92–0.96).
Table 2.
VOC-based exhaled breath and urine tests for the diagnosis of different types of tumors.
| Sensitivity (95% CI) | Specificity (95% CI) | PLR (95% CI) | NLR (95% CI) | AUC (95% CI) | |
|---|---|---|---|---|---|
| Overall (n=90) | 0.89 (0.87–0.91) | 0.88 (0.85–0.90) | 7.4 (6.0–9.1) | 0.13 (0.10–0.15) | 0.95 (0.92–0.96) |
| Lung cancer (n=38) | 0.91 (0.88–0.93) | 0.90 (0.86–0.93) | 9.0 (6.4–12.8) | 0.10 (0.07–0.14) | 0.96 (0.94–0.97) |
| Colorectal cancer (n=11) | 0.87 (0.82–0.91) | 0.80 (0.70–0.87) | 4.3 (2.8–6.7) | 0.16 (0.11–0.23) | 0.91 (0.88–0.93) |
| Urologic neoplasms (n=9) | 0.81 (0.76–0.85) | 0.86 (0.81–0.91) | 6.0 (4.1–8.7) | 0.22 (0.17–0.28) | 0.89 (0.86–0.91) |
| Breast cancer (n=8) | 0.91 (0.79–0.96) | 0.90 (0.67–0.97) | 8.8 (2.3–34.0) | 0.10 (0.04–0.28) | 0.96 (0.94–0.97) |
| Gastroesophageal cancer (n=7) | 0.92 (0.80–0.97) | 0.92 (0.84–0.97) | 12.0 (5.3–27.1) | 0.09 (0.03–0.23) | 0.97 (0.95–0.98) |
| Head and neck cancer (n=7) | 0.86 (0.79–0.91) | 0.78 (0.71–0.84) | 3.9 (2.9–5.4) | 0.17 (0.11–0.28) | 0.88 (0.84–0.90) |
| Hepatocellular carcinoma (n=4) | 0.72 (0.65–0.78) | 0.86 (0.79–0.91) | 5.1 (3.4–7.8) | 0.32 (0.25–0.42) | 0.83 (0.80–0.86) |
AUC, area under the curve; NLR, negative likelihood ratio; PLR, positive likelihood ratio.
Figure 5.
Fagan nomograms for the elucidation of post-test probabilities with different pretest probabilities. (A) The pretest probability was 0.25, yielding a PPV of 71% and a NPV of 4%. (B) The pretest probability was 0.51, yielding a PPV of 88% and a NPV of 12%. (C) The pretest probability was 0.75, yielding a PPV of 96% and a NPV of 28%.
Univariable meta-regression and subgroup analysis
In order to investigate the origins of heterogeneity, we conducted meta-regression and subgroup analysis across six dimensions, as depicted in Figure 6 and Table 3. These dimensions encompassed the nation of the included patients (developing country vs. developed country), tumor type (lung cancer vs. other cancers), control group (health controls vs. other controls), sample size (<80 vs. >80 patients), test sample (VOCs from exhaled breath or urine), and analytical platform (e-Nose vs. non-e-Nose). The findings indicated that all the six parameters exhibited heterogeneity in both sensitivity and specificity (P<0.05). The subgroup analysis, as presented in Table 3, provided a summary of the results. Notably, studies conducted in developing countries, focusing on lung cancer, utilizing health controls, involving fewer than 80 patients, utilizing exhaled breath samples, and employing e-Nose demonstrated higher pooled sensitivities and specificities compared to studies conducted in developed countries, involving other types of cancers, utilizing other control groups, including more than 80 patients, utilizing urine samples, and not utilizing e-Nose technology.
Figure 6.
Univariable meta-regression and subgroup analysis for differentiating cancers.
Table 3.
Univariable meta-regression and subgroup analysis for differentiating cancers.
| Parameter | Category | No. of studies | Sensitivity (95% CI) | P | Specificity (95% CI) | P |
|---|---|---|---|---|---|---|
| Nation | Developing country | 31 | 0.92 (0.90–0.95) | <0.001 | 0.92 (0.89–0.95) | <0.001 |
| Developed country | 59 | 0.86 (0.84–0.89) | 0.85 (0.82–0.88) | |||
| Tumor type | Lung cancer | 38 | 0.91 (0.88–0.93) | <0.001 | 0.90 (0.86–0.93) | <0.001 |
| Other cancers | 52 | 0.87 (0.84–0.90) | 0.87 (0.83–0.90) | |||
| Control group | Health controls | 51 | 0.90 (0.88–0.92) | <0.001 | 0.90 (0.87–0.93) | <0.001 |
| Other controls | 39 | 0.87 (0.83–0.90) | 0.85 (0.81–0.89) | |||
| Sample size | <80 | 32 | 0.89 (0.86–0.93) | <0.001 | 0.91 (0.88–0.95) | <0.001 |
| >80 | 58 | 0.89 (0.86–0.91) | 0.86 (0.83–0.89) | |||
| Test sample | Exhaled breath | 69 | 0.89 (0.87–0.92) | <0.001 | 0.88 (0.85–0.91) | <0.001 |
| Urine | 21 | 0.87 (0.82–0.91) | 0.88 (0.83–0.93) | |||
| Analytical platform | e-Nose | 41 | 0.91 (0.88–0.93) | <0.001 | 0.89 (0.85–0.92) | <0.001 |
| non-e-Nose | 49 | 0.87 (0.84–0.90) | 0.87 (0.84–0.91) |
Origin analysis and origin-based pathway analysis
Constrained by the limited number of compounds, we conducted a focused investigation into the metabolic pathways of four specific types of tumors, namely lung, colorectal, breast, and liver cancers. Additionally, we included relevant studies on VOCs in colorectal and breast cancers88,104–116. Following the removal of not-found and duplicate compounds, we identified 79, 102, 49, and 42 unique VOCs associated with lung cancer, colorectal cancer, breast cancer, and hepatocellular carcinoma, respectively (Supplementary Table S2, Supplemental Digital Content 5, http://links.lww.com/JS9/B590). These metabolites were deemed to exhibit significant alterations.
The metabolites of lung cancer were initially derived from various sources, including four metabolites specific to the human host, 45 metabolites originating from bacteria, and 134 metabolites from other sources such as drugs, food, the environment, and unknown origins (Supplementary Fig. S1A, Supplemental Digital Content 6, http://links.lww.com/JS9/B591). In Supplementary Figs S1B-C (Supplemental Digital Content 6, http://links.lww.com/JS9/B591), we compared the total number of metabolites and metabolic pathways belonging to the host with those belonging to the microbiota. A total of 14 metabolites (isopropanol, butane, n-butanal, butanol, methanol, methylacetate, acetone, 2,3-butandione, cyclohexane, cyclohexanone, toluene, benzaldehyde, propanal, and o-xylene) were identified as significantly specific to microbiota in four enriched metabolic pathways (butanoate metabolism (ko00650), caprolactam degradation (ko00930), toluene degradation (ko00623), and xylene degradation (ko00622)) (Table 4; Supplementary Fig. S1D, Supplemental Digital Content 6, http://links.lww.com/JS9/B591). The host metabolites did not have an enriched metabolic pathway, whereas 10 significantly metabolic pathways were enriched in the human and microbial communities (Supplementary Table S3, Supplemental Digital Content 7, http://links.lww.com/JS9/B592; Supplementary Fig. S1D, Supplemental Digital Content 6, http://links.lww.com/JS9/B591).
Table 4.
Significantly metabolic pathways enriched in the microbial communities.
| Pathway name | Pathway ID | Sig. volatile metabolites | P | |
|---|---|---|---|---|
| Lung cancer | Butanoate metabolism | ko00650 | Isopropanol; Butane; n-Butanal; Butanol; Methanol; Methylacetate; Acetone; 2,3-Butandione | 8.21e-11 |
| Caprolactam degradation | ko00930 | Cyclohexane; Cyclohexanone | 4.82e-03 | |
| Toluene degradation | ko00623 | Toluene; Benzaldehyde | 2.19e-02 | |
| Xylene degradation | ko00622 | Propanal; o-Xylene | 2.30e-02 | |
| Colorectal cancer | Toluene degradation | ko00623 | Methylbenzene; Benzaldehyde; p-Cresol | 1.25e-03 |
| Xylene degradation | ko00622 | m-Cymene; p-Xylene; p-Cymene | 1.35e-03 | |
| Butanoate metabolism | ko00650 | 2,3-Butanedione; Butanal; Acetone | 2.07e-03 | |
| Ethylbenzene degradation | ko00642 | Ethylbenzene; 1-Phenylethanone | 2.96e-03 | |
| Dioxin degradation | ko00621 | Biphenyl; Dibenzofuran | 1.60e-02 | |
| Biosynthesis of various alkaloids | ko00996 | Vanillin | 2.37e-02 | |
| Breast cancer | Butanoate metabolism | ko00650 | Methanol; 2-Propanol; Acetone | 1.69e-04 |
| Furfural degradation | ko00365 | Furfural | 2.13e-02 | |
| Ethylbenzene degradation | ko00642 | Ethylbenzene | 2.71e-02 | |
| Caprolactam degradation | ko00930 | Cyclohexanone | 4.48e-02 | |
| Hepatocellular carcinoma | Butanoate metabolism | ko00650 | Acetone; 2-Propanol; Butanal; 2,3-Butandione; 1-Butanol | 2.30e-07 |
| Xylene degradation | ko00622 | m-Xylene; p-Xylene | 7.71e-03 |
The initial origins of colorectal cancer metabolites encompassed various sources, including 35 host metabolites, 74 bacterial metabolites, and 179 metabolites from other origins such as drugs, food, environment, and unknown sources (Supplementary Fig. S2A, Supplemental Digital Content 8, http://links.lww.com/JS9/B593). Supplementary Figs S2B–C, (Supplemental Digital Content 8, http://links.lww.com/JS9/B593) illustrated the total number of metabolites and metabolic pathways attributed to the host, and provide a comparison with those associated with the microbiota. A total of 14 metabolites (methylbenzene, benzaldehyde, p-cresol, m-cymene, p-xylene, p-cymene, 2,3-butanedione, butanal, acetone, ethylbenzene, 1-phenylethanone, biphenyl, dibenzofuran, and vanillin) were identified across six enriched metabolic pathways, namely toluene degradation (ko00623), xylene degradation (ko00622), butanoate metabolism (ko00650), ethylbenzene degradation (ko00642), dioxin degradation (ko00621), and biosynthesis of various alkaloids (ko00996) (Table 4; Supplementary Fig. S2D, Supplemental Digital Content 8, http://links.lww.com/JS9/B593). These metabolites exhibited a significant association with the microbiota. Conversely, no enriched metabolic pathway specific to host metabolites was observed. Additionally, a total of 37 significantly metabolic pathways were found to be enriched in both human and microbial communities (Supplementary Table S3, Supplemental Digital Content 7, http://links.lww.com/JS9/B592; Supplementary Fig. S2D, Supplemental Digital Content 8, http://links.lww.com/JS9/B593).
The metabolites associated with breast cancer were initially derived from various sources, including 12 host metabolites, 32 bacterial metabolites, and 90 others originating from drugs, food, the environment, and unknown origins (Supplementary Fig. S3A, Supplemental Digital Content 9, http://links.lww.com/JS9/B594). Supplementary Figs. S3B–C (Supplemental Digital Content 9, http://links.lww.com/JS9/B594) illustrated the comprehensive count of metabolites and metabolic pathways associated with the host, juxtaposed with those of the microbiota. The microbiota exhibited a noteworthy specificity, with six metabolites (methanol, 2-propanol, acetone, furfural, ethylbenzene, and cyclohexanone) distributed across four enriched metabolic pathways (namely, butanoate metabolism (ko00650), furfural degradation (ko00365), ethylbenzene degradation (ko00642), and caprolactam degradation (ko00930)) (Table 4; Supplementary Fig. S3D, Supplemental Digital Content 9, http://links.lww.com/JS9/B594). Conversely, no enriched metabolic pathways were identified for host metabolites, while five significantly metabolic pathways demonstrated enrichment in both human and microbial communities (Supplementary Table S3, Supplemental Digital Content 7, http://links.lww.com/JS9/B592; Supplementary Fig. S3D, Supplemental Digital Content 9, http://links.lww.com/JS9/B594).
The metabolites of hepatocellular carcinoma were initially derived from various sources, including four host metabolites, 25 bacterial metabolites, and 72 others (such as drugs, food, environment, and unknown sources) (Supplementary Fig. S4A, Supplemental Digital Content 10, http://links.lww.com/JS9/B595). Supplementary Figs S4B–C, (Supplemental Digital Content 10, http://links.lww.com/JS9/B595) illustrated the total number of metabolites and metabolic pathways associated with the host and compare them to those of the microbiota. Among these, seven metabolites (acetone, 2-propanol, butanal, 2,3-butandione, 1-butanol, m-xylene, and p-xylene) in two enriched metabolic pathways (butanoate metabolism (ko00650) and xylene degradation (ko00622)) were found to be significantly specific to the microbiota (Table 4; Supplementary Fig. S4D, Supplemental Digital Content 10, http://links.lww.com/JS9/B595). No enriched metabolic pathway was identified for host metabolites, while eight significantly metabolic pathways were enriched in both human and microbiota (Supplementary Table S3, Supplemental Digital Content 7, http://links.lww.com/JS9/B592; Supplementary Fig. S4D, Supplemental Digital Content 10, http://links.lww.com/JS9/B595).
Discussion
Over the past decade, numerous proof-of-principle studies have provided evidence for the effectiveness of employing VOC profiles in clinical diagnostics. Notably, VOC analysis has proven successful in detecting various cancers, including lung, breast, prostate, gastric, colorectal, and liver cancers. While further validation through larger-scale studies is necessary, these findings appear to corroborate the notion that pathological mechanisms within the body can impact VOC profiles, thereby yielding distinct VOC signatures for each disease. The objective of the present study was to conduct a comprehensive review and meta-analysis of prior research investigating the use of exhaled or urinary VOCs for the purpose of cancer detection. In addition to their role as indicators of tumor metabolism, this study also aimed to investigate the metabolic pathways associated with the development of tumors.
The analysis of VOCs in exhaled breath and urine presented a promising approach for the diagnosis of various tumor types. Our study demonstrated the efficacy of VOC analysis in exhaled breath and urine for cancer diagnosis, as evidenced by an area under the receiver operating characteristic curve of 0.95 (95% CI: 0.92–0.96), a pooled sensitivity of 0.89 (95% CI: 0.87–0.91), and a pooled specificity of 0.88 (95% CI: 0.85–0.90). These parameters collectively suggested that this test showed superior diagnostic performance in distinguishing tumors from control groups. The diagnostic accuracy of LRs (PLR and NLR) is widely acknowledged in academic circles, particularly for their utility in clinical decision-making, surpassing sensitivity, specificity, or AUC. To be deemed highly useful, a diagnostic test should possess a PLR exceeding 10.0 and an NLR below 0.10, as recommended by a commonly employed heuristic for LR interpretation117. A higher PLR suggests proficiency in disease diagnosis, whereas a lower NLR suggests proficiency in disease exclusion. The PLR and NLR of VOCs in cancer diagnosis were determined to be 7.4 and 0.13, respectively. These findings indicate that VOCs exhibit a greater ability to rule out tumor diagnosis, while their capacity to confirm it is comparatively lower.
Additionally, we have employed Fagan’s nomogram to leverage the findings from our diagnostic analysis for the purpose of estimating the likelihood of cancer in patients during routine clinical practice118. According to the findings depicted in Figure 5, patients exhibiting suspected cancer with a pretest probability of 25% experienced a notable increase in the likelihood of cancer to 77% upon receiving a positive test result. Conversely, a negative test result led to a reduction in the probability to 6%. Similarly, patients with suspected cancer and a pretest probability of 75% observed a substantial rise in the likelihood to 97% following a positive test result, while a negative test result resulted in a decrease to 37% probability. When evaluating a higher pretest probability, it was found that the ‘ruling in’ probability of cancer was more suitable than ‘ruling out’. Conversely, a lower pretest probability increased the probability of cancer for ‘ruling out’. The pretest probability was notably established at 51%, while the likelihood of developing cancer following a positive result was determined to be 88%. Stated differently, the probability of not having cancer subsequent to a positive result was 12%, which corresponded to the probability of being cancer-free after a negative result. This indicated that VOCs achieved comparable performance in both confirming and excluding the disease. Hence, a pretest probability of 51% can be considered as the threshold for diagnosing cancers with VOCs. As the estimated pretest probability of cancer exceeds 51%, it becomes more appropriate to emphasize the ‘ruling in’ approach. Conversely, when the estimated pretest probability of cancer falls below 51%, it is more suitable to emphasize the ‘ruling out’ approach.
In order to ascertain potential sources of heterogeneity, we conducted univariate meta-regression and meta-analysis. These analyses unveiled that various factors, such as nation, tumor type, sample size, source of VOCs, analytical platforms, and types of the control group, may contribute to the heterogeneity observed in our meta-analysis. Further analysis demonstrated a higher level of precision in detecting VOCs for cancer diagnosis in developing countries in contrast to developed countries, suggesting that the effectiveness of VOCs in cancer screening might vary depending on racial or ethnic factors. When considering the detection of VOCs, the diagnostic efficacy of utilizing urine was equally optimal in comparison to exhaled breath. The utilization of urinary VOC analysis in clinical tumor diagnosis practice offered distinct advantages over exhaled VOC analysis, thus warranting further investigation into the potential of urinary VOCs. In the context of the analytical platform, the utilization of e-Nose for the analysis of VOCs exhibited superior sensitivity and specificity in comparison to alternative platforms. Due to the heightened sensitivity of e-Noses toward alterations in both endogenous and exogenous factors, the diagnostic precision may exhibit variability across diverse research settings and patient cohorts119. Consequently, it becomes imperative to undertake extensive, multicenter external validation studies to explore the applicability and replicability of the findings across various research settings and patient populations.
The microbiome and its metabolites exhibit a strong correlation with human health and diseases. Present research predominantly employs statistical correlation analysis to investigate the interplay between microbial communities and all identified metabolites, thereby elucidating their association120. It is of great importance to undertake additional measures in order to distinguish the metabolic activities attributed to the host, bacteria, or both. Otherwise, comprehending the genuine metabolic functions of microbial metabolites becomes arduous when all metabolites from the host and bacterial communities are combined. Differentiating between microbial metabolites and other sources such as the host, food, or environment, and investigating their metabolic functions and associations with the microbiome, could potentially enhance the effectiveness and precision of biomarker identification. The second aim of the current study was to investigate the metabolic pathways linked to the development of cancerous tumors, as VOCs serve as markers for both tumor presence and human metabolism. We identified a total of 14, 14, 6, and 7 VOCs associated with microbiota in lung, colorectal, breast, and liver cancers, respectively. The enrichment analysis of these volatile metabolites demonstrated a significant enrichment of butanoate metabolism in the aforementioned types of tumors. Butyric acid, a significant metabolite generated through the fermentation of intestinal flora, serves as a transport substrate in butanoate metabolism and plays a crucial role in regulating host energy homeostasis mediated by intestinal flora. Recent literature has highlighted the potential impact of butyrate on the development of diverse diseases, including cancer. Sodium butyrate has been observed to enhance the expression levels of P-gp and signal transducer and activator of transcription (STAT) 3, as well as promoted STAT3 phosphorylation and enhanced mRNA stability of ATP-binding cassette subfamily B member 1 in human lung cancer cells121. Zhang et al.122 indicated that butyrate induced apoptosis through the activation of mitogen-activated protein kinase signaling in colon cancer RKO cells. Sodium butyrate elicited apoptosis through the augmentation of reactive oxygen species levels, enhancement of caspase activity, and reduction of mitochondrial membrane potential in both normal breast and breast cancer cells123. Butyrate exerted inhibitory effects on cell proliferation and induced apoptosis in both Hep3B and HepG2 cells. Prolonged exposure to butyrate was associated with enhanced hepatocyte growth and fibrosis in the liver. Therefore, butyrate significantly impacted the proliferation of liver cells124.
Conclusions
The analysis of exhaled breath and urinary VOCs, particularly utilizing e-Nose technology, showed promise for cancer screening. However, it was imperative to conduct multicenter validation studies to confirm its efficacy, as well as to evaluate the performance of the optimal analytical approach and consider confounding factors. For example, before clinical implementation can be realized, the lack of standardization and reproducibility in the field of e-Nose research must be addressed. In addition, the enrichment analysis of volatile metabolites revealed a significant enrichment of butanoate metabolism in four tumor types, namely lung, colorectum, breast, and liver. These findings hold significant implications for the prospective clinical application of multiomics correlation in disease management and the exploration of potential therapeutic targets.
Ethical approval
The article is a systematic review and does not require ethical approval.
Consent
The article is a systematic review and does not require ethics committee approval and fully informed written consent.
Sources of funding
This work was supported by the Natural Science Foundation of Qinghai Province of China (No. 2022-ZJ-912).
Author contribution
M.Z., Q.H.W., X.Y.L., Y.C., J.Z.X., and D.Z.C.: study design; M.Z., Q.H.W., X.Y.L., P.Z., and R.Y.: systematic literature search; M.Z., Q.H.W., and X.Y.L.: collected data and drafted the manuscript; M.Z., Q.H.W., X.Y.L., and Y.C.: data analysis and interpretation; M.Z., Q.H.W., X.Y.L., and Y.C.: statistical analysis; M.Z., Q.H.W., X.Y.L., and Y.C.: manuscript preparation. All authors critically revised the manuscript for important intellectual content. All authors had full access to all the data and approved the final version of the manuscript, including the authorship list.
Conflicts of interest disclosure
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Research registration unique identifying number (UIN)
Research registry registration number: reviewregistry 1702.
Guarantor
Daozhen Chen is the guarantor of this article.
Data availability statement
The main data supporting the findings of this study are included within the article and its supplementary materials. Additional data are available from the corresponding authors upon reasonable request.
Provenance and peer review
Not commissioned, externally peer-reviewed.
Supplementary Material
Supplementary Material

Footnotes
M.Z., Q.W., and X.L. contributed equally to this work.
Sponsorships or competing interests that may be relevant to content are disclosed at the end of this article.
Supplemental Digital Content is available for this article. Direct URL citations are provided in the HTML and PDF versions of this article on the journal's website, www.lww.com/international-journal-of-surgery.
Published online 18 December 2023
Contributor Information
Min Zhou, Email: zhoumin1119@foxmail.com.
Qinghua Wang, Email: qinghua970406@163.com.
Xinyi Lu, Email: luxinyi0812@163.com.
Ping Zhang, Email: zsyl2008@163.com.
Rui Yang, Email: yangrui826@126.com.
Yu Chen, Email: cy-78@hotmail.com.
Jiazeng Xia, Email: jiazengxia@yahoo.com.
Daozhen Chen, Email: chendaozhen@163.com.
References
- 1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin 2022;72:7–33. [DOI] [PubMed] [Google Scholar]
- 2. Koo MM, Swann R, McPhail S, et al. Presenting symptoms of cancer and stage at diagnosis: evidence from a cross-sectional, population-based study. Lancet Oncol 2020;21:73–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Newgard CB. Metabolomics and metabolic diseases: where do we stand? Cell Metab 2017;25:43–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Lange J, Eddhif B, Tarighi M, et al. Volatile organic compound based probe for induced volatolomics of cancers. Angew Chem Int Ed Engl 2019;58:17563–17566. [DOI] [PubMed] [Google Scholar]
- 5. van Liere ELSA, van Dijk LJ, Bosch S, et al. Urinary volatile organic compounds for colorectal cancer screening: A systematic review and meta-analysis. Eur J Cancer 2023;186:69–82. [DOI] [PubMed] [Google Scholar]
- 6. Janssens E, van Meerbeeck JP, Lamote K. Volatile organic compounds in human matrices as lung cancer biomarkers: a systematic review. Crit Rev Oncol Hematol 2020;153:103037. [DOI] [PubMed] [Google Scholar]
- 7. Lubes G, Goodarzi M. GC-MS based metabolomics used for the identification of cancer volatile organic compounds as biomarkers. J Pharm Biomed Anal 2018;147:313–322. [DOI] [PubMed] [Google Scholar]
- 8. Khatoon Z, Fouad H, Alothman OY, et al. Doped SnO2 nanomaterials for e-Nose based electrochemical sensing of biomarkers of lung cancer. ACS Omega 2020;5:27645–27654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Yakubu HG, Kovacs Z, Toth T, et al. Trends in artificial aroma sensing by means of electronic nose technologies to advance dairy production - a review. Crit Rev Food Sci Nutr 2023;63:234–248. [DOI] [PubMed] [Google Scholar]
- 10. Hintzen KFH, Grote J, Wintjens AGWE, et al. Breath analysis for the detection of digestive tract malignancies: systematic review. BJS Open 2021;5:zrab013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Drabińska N, Flynn C, Ratcliffe N, et al. A literature survey of all volatiles from healthy human breath and bodily fluids: the human volatilome. J Breath Res 2021;15:034001. doi: 10.1088/1752-7163/abf1d0 [DOI] [PubMed] [Google Scholar]
- 12. Tsou PH, Lin ZL, Pan YC, et al. Exploring volatile organic compounds in breath for high-accuracy prediction of lung cancer. Cancers (Basel) 2021;13:1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Becker R. Non-invasive cancer detection using volatile biomarkers: is urine superior to breath? Med Hypotheses 2020;143:110060. [DOI] [PubMed] [Google Scholar]
- 14. Page MJ, McKenzie JE, Bossuyt PM, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Int J Surg 2021;88:105906. [DOI] [PubMed] [Google Scholar]
- 15. Shea BJ, Reeves BC, Wells G, et al. AMSTAR 2: a critical appraisal tool for systematic reviews that include randomised or non-randomised studies of healthcare interventions, or both. BMJ 2017;358:j4008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Yu G, Xu C, Zhang D, et al. MetOrigin: discriminating the origins of microbial metabolites for integrative analysis of the gut microbiome and metabolome. iMeta 2022;1:e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Moses LE, Shapiro D, Littenberg B. Combining independent studies of a diagnostic test into a summary ROC curve: data-analytic approaches and some additional considerations. Stat Med 1993;12:1293–1316. [DOI] [PubMed] [Google Scholar]
- 18. Deeks JJ. Systematic reviews in health care: systematic reviews of evaluations of diagnostic and screening tests. BMJ 2001;323:157–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Phillips M, Gleeson K, Hughes JM, et al. Volatile organic compounds in breath as markers of lung cancer: a cross-sectional study. Lancet 1999;353:1930–1933. [DOI] [PubMed] [Google Scholar]
- 20. Phillips M, Cataneo RN, Cummin AR, et al. Detection of lung cancer with volatile markers in the breath. Chest 2003;123:2115–2123. [DOI] [PubMed] [Google Scholar]
- 21. Phillips M, Cataneo RN, Ditkoff BA, et al. Volatile markers of breast cancer in the breath. Breast J 2003;9:184–191. [DOI] [PubMed] [Google Scholar]
- 22. Machado RF, Laskowski D, Deffenderfer O, et al. Detection of lung cancer by sensor array analyses of exhaled breath. Am J Respir Crit Care Med 2005;171:1286–1291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Phillips M, Altorki N, Austin JH, et al. Detection of lung cancer using weighted digital analysis of breath biomarkers. Clin Chim Acta 2008;393:76–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Ligor M, Ligor T, Bajtarevic A, et al. Determination of volatile organic compounds in exhaled breath of patients with lung cancer using solid phase microextraction and gas chromatography mass spectrometry. Clin Chem Lab Med 2009;47:550–560. [DOI] [PubMed] [Google Scholar]
- 25. Song G, Qin T, Liu H, et al. Quantitative breath analysis of volatile organic compounds of lung cancer patients. Lung Cancer 2010;67:227–231. [DOI] [PubMed] [Google Scholar]
- 26. Qin T, Liu H, Song Q, et al. The screening of volatile markers for hepatocellular carcinoma. Cancer Epidemiol Biomarkers Prev 2010;19:2247–2253. [DOI] [PubMed] [Google Scholar]
- 27. Phillips M, Cataneo RN, Saunders C, et al. Volatile biomarkers in the breath of women with breast cancer. J Breath Res 2010;4:026003. [DOI] [PubMed] [Google Scholar]
- 28. Dragonieri S, van der Schee MP, Massaro T, et al. An electronic nose distinguishes exhaled breath of patients with malignant pleural mesothelioma from controls. Lung Cancer 2012;75:326–331. [DOI] [PubMed] [Google Scholar]
- 29. Peled N, Hakim M, Bunn PA, Jr, et al. Non-invasive breath analysis of pulmonary nodules. J Thorac Oncol 2012;7:1528–1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Chapman EA, Thomas PS, Stone E, et al. A breath test for malignant mesothelioma using an electronic nose. Eur Respir J 2012;40:448–454. [DOI] [PubMed] [Google Scholar]
- 31. Altomare DF, Di Lena M, Porcelli F, et al. Exhaled volatile organic compounds identify patients with colorectal cancer. Br J Surg 2013;100:144–150. [DOI] [PubMed] [Google Scholar]
- 32. Broza YY, Kremer R, Tisch U, et al. A nanomaterial-based breath test for short-term follow-up after lung tumor resection. Nanomedicine 2013;9:15–21. [DOI] [PubMed] [Google Scholar]
- 33. Xu ZQ, Broza YY, Ionsecu R, et al. A nanomaterial-based breath test for distinguishing gastric cancer from benign gastric conditions. Br J Cancer 2013;108:941–950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Handa H, Usuba A, Maddula S, et al. Exhaled breath analysis for lung cancer detection using ion mobility spectrometry. PLoS One 2014;9:e114555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Arasaradnam RP, McFarlane MJ, Ryan-Fisher C, et al. Detection of colorectal cancer (CRC) by urinary volatile organic compound analysis. PLoS One 2014;9:e108750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Bousamra M, II, Schumer E, Li M, et al. Quantitative analysis of exhaled carbonyl compounds distinguishes benign from malignant pulmonary disease. J Thorac Cardiovasc Surg 2014;148:1074–1081. [DOI] [PubMed] [Google Scholar]
- 37. Gruber M, Tisch U, Jeries R, et al. Analysis of exhaled breath for diagnosing head and neck squamous cell carcinoma: a feasibility study. Br J Cancer 2014;111:790–798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Fu XA, Li M, Knipp RJ, et al. Noninvasive detection of lung cancer using exhaled breath. Cancer Med 2014;3:174–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Leunis N, Boumans ML, Kremer B, et al. Application of an electronic nose in the diagnosis of head and neck cancer. Laryngoscope 2014;124:1377–1381. [DOI] [PubMed] [Google Scholar]
- 40. Schumer EM, Trivedi JR, van Berkel V, et al. High sensitivity for lung cancer detection using analysis of exhaled carbonyl compounds. J Thorac Cardiovasc Surg 2015;150:1517–1524. [DOI] [PubMed] [Google Scholar]
- 41. Li M, Yang D, Brock G, et al. Breath carbonyl compounds as biomarkers of lung cancer. Lung Cancer 2015;90:92–97. [DOI] [PubMed] [Google Scholar]
- 42. Ligor T, Pater Ł, Buszewski B. Application of an artificial neural network model for selection of potential lung cancer biomarkers. J Breath Res 2015;9:027106. [DOI] [PubMed] [Google Scholar]
- 43. Kumar S, Huang J, Abbassi-Ghadi N, et al. Mass spectrometric analysis of exhaled breath for the identification of volatile organic compound biomarkers in esophageal and gastric adenocarcinoma. Ann Surg 2015;262:981–990. [DOI] [PubMed] [Google Scholar]
- 44. Amal H, Shi DY, Ionescu R, et al. Assessment of ovarian cancer conditions from exhaled breath. Int J Cancer 2015;136:E614–E622. [DOI] [PubMed] [Google Scholar]
- 45. Guo L, Wang C, Chi C, et al. Exhaled breath volatile biomarker analysis for thyroid cancer. Transl Res 2015;166:188–195. [DOI] [PubMed] [Google Scholar]
- 46. Barash O, Zhang W, Halpern JM, et al. Differentiation between genetic mutations of breast cancer by breath volatolomics. Oncotarget 2015;6:44864–44876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Rocco R, Incalzi RA, Pennazza G, et al. BIONOTE e-nose technology may reduce false positives in lung cancer screening programmes†. Eur J Cardiothorac Surg 2016;49:1112–1117. [DOI] [PubMed] [Google Scholar]
- 48. Amal H, Leja M, Funka K, et al. Breath testing as potential colorectal cancer screening tool. Int J Cancer 2016;138:229–236. [DOI] [PubMed] [Google Scholar]
- 49. Amal H, Leja M, Funka K, et al. Detection of precancerous gastric lesions and gastric cancer through exhaled breath. Gut 2016;65:400–407. [DOI] [PubMed] [Google Scholar]
- 50. Schallschmidt K, Becker R, Jung C, et al. Comparison of volatile organic compounds from lung cancer patients and healthy controls-challenges and limitations of an observational study. J Breath Res 2016;10:046007. [DOI] [PubMed] [Google Scholar]
- 51. Zou X, Zhou W, Lu Y, et al. Exhaled gases online measurements for esophageal cancer patients and healthy people by proton transfer reaction mass spectrometry. J Gastroenterol Hepatol 2016;31:1837–1843. [DOI] [PubMed] [Google Scholar]
- 52. Gasparri R, Santonico M, Valentini C, et al. Volatile signature for the early diagnosis of lung cancer. J Breath Res 2016;10:016007. [DOI] [PubMed] [Google Scholar]
- 53. Sakumura Y, Koyama Y, Tokutake H, et al. Diagnosis by volatile organic compounds in exhaled breath from lung cancer patients using support vector machine algorithm. Sensors (Basel) 2017;17:287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Li W, Liu H, Xie D, et al. Lung cancer screening based on type-different sensor arrays. Sci Rep 2017;7:1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Shlomi D, Abud M, Liran O, et al. Detection of lung cancer and EGFR mutation by electronic nose system. J Thorac Oncol 2017;12:1544–1551. [DOI] [PubMed] [Google Scholar]
- 56. Schuermans VNE, Li Z, Jongen ACHM, et al. Pilot study: detection of gastric cancer from exhaled air analyzed with an electronic nose in Chinese patients. Surg Innov 2018;25:429–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Heers H, Gut JM, Hegele A, et al. Non-invasive detection of bladder tumors through volatile organic compounds: a pilot study with an electronic nose. Anticancer Res 2018;38:833–837. [DOI] [PubMed] [Google Scholar]
- 58. Markar SR, Brodie B, Chin ST, et al. Profile of exhaled-breath volatile organic compounds to diagnose pancreatic cancer. Br J Surg 2018;105:1493–1500. [DOI] [PubMed] [Google Scholar]
- 59. Widlak MM, Neal M, Daulton E, et al. Risk stratification of symptomatic patients suspected of colorectal cancer using faecal and urinary markers. Colorectal Dis 2018;20:O335–O342. [DOI] [PubMed] [Google Scholar]
- 60. Marzorati D, Mainardi L, Sedda G, et al. A metal oxide gas sensors array for lung cancer diagnosis through exhaled breath analysis. Annu Int Conf IEEE Eng Med Biol Soc 2019;2019:1584–1587. [DOI] [PubMed] [Google Scholar]
- 61. Nissinen SI, Roine A, Hokkinen L, et al. Detection of pancreatic cancer by urine volatile organic compound analysis. Anticancer Res 2019;39:73–79. [DOI] [PubMed] [Google Scholar]
- 62. Rudnicka J, Kowalkowski T, Buszewski B. Searching for selected VOCs in human breath samples as potential markers of lung cancer. Lung Cancer 2019;135:123–129. [DOI] [PubMed] [Google Scholar]
- 63. Chandran D, Ooi EH, Watson DI, et al. The use of selected ion flow tube-mass spectrometry technology to identify breath volatile organic compounds for the detection of head and neck squamous cell carcinoma: a pilot study. Medicina (Kaunas) 2019;55:306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Mozdiak E, Wicaksono AN, Covington JA, et al. Colorectal cancer and adenoma screening using urinary volatile organic compound (VOC) detection: early results from a single-centre bowel screening population (UK BCSP). Tech Coloproctol 2019;23:343–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Mohamed EI, Mohamed MA, Abdel-Mageed SM, et al. Volatile organic compounds of biofluids for detecting lung cancer by an electronic nose based on artificial neural network. J Appl Biomed 2019;17:67. [DOI] [PubMed] [Google Scholar]
- 66. Díaz de León-Martínez L, Rodríguez-Aguilar M, Gorocica-Rosete P, et al. Identification of profiles of volatile organic compounds in exhaled breath by means of an electronic nose as a proposal for a screening method for breast cancer: a case-control study. J Breath Res 2020;14:046009. [DOI] [PubMed] [Google Scholar]
- 67. Chen Q, Chen Z, Liu D, et al. Constructing an e-Nose using metal-ion-induced assembly of graphene oxide for diagnosis of lung cancer via exhaled breath. ACS Appl Mater Interfaces 2020;12:17713–17724. [DOI] [PubMed] [Google Scholar]
- 68. van Keulen KE, Jansen ME, Schrauwen RWM, et al. Volatile organic compounds in breath can serve as a non-invasive diagnostic biomarker for the detection of advanced adenomas and colorectal cancer. Aliment Pharmacol Ther 2020;51:334–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Waltman CG, Marcelissen TAT, van Roermund JGH. Exhaled-breath testing for prostate cancer based on volatile organic compound profiling using an electronic nose device (Aeonose™): a preliminary report. Eur Urol Focus 2020;6:1220–1225. [DOI] [PubMed] [Google Scholar]
- 70. Kort S, Tiggeloven MM, Brusse-Keizer M, et al. Multi-centre prospective study on diagnosing subtypes of lung cancer by exhaled-breath analysis. Lung Cancer 2018;125:223–229. [DOI] [PubMed] [Google Scholar]
- 71. Altomare DF, Picciariello A, Rotelli MT, et al. Chemical signature of colorectal cancer: case-control study for profiling the breath print. BJS Open 2020;4:1189–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Gharra A, Broza YY, Yu G, et al. Exhaled breath diagnostics of lung and gastric cancers in China using nanosensors. Cancer Commun (Lond) 2020;40:273–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Zhu S, Huang Z, Nabi G. Fluorometric optical sensor arrays for the detection of urinary bladder cancer specific volatile organic compounds in the urine of patients with frank hematuria: a prospective case-control study. Biomed Opt Express 2020;11:1175–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Kort S, Brusse-Keizer M, Gerritsen JW, et al. Improving lung cancer diagnosis by combining exhaled-breath data and clinical parameters. ERJ Open Res 2020;6:00221-2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Tyagi H, Daulton E, Bannaga AS, et al. Urinary volatiles and chemical characterisation for the non-invasive detection of prostate and bladder cancers. Biosensors (Basel) 2021;11:437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Capelli L, Bax C, Grizzi F, et al. Optimization of training and measurement protocol for eNose analysis of urine headspace aimed at prostate cancer diagnosis. Sci Rep 2021;11:20898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Tyagi H, Daulton E, Bannaga AS, et al. Non-invasive detection and staging of colorectal cancer using a portable electronic nose. Sensors (Basel) 2021;21:5440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Yang HY, Wang YC, Peng HY, et al. Breath biopsy of breast cancer using sensor array signals and machine learning analysis. Sci Rep 2021;11:103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Hong Y, Che X, Su H, et al. Exhaled breath analysis using on-line preconcentration mass spectrometry for gastric cancer diagnosis. J Mass Spectrom 2021;56:e4588. [DOI] [PubMed] [Google Scholar]
- 80. B VA, Subramoniam M, Mathew L. Detection of COPD and lung cancer with electronic nose using ensemble learning methods. Clin Chim Acta 2021;523:231–238. [DOI] [PubMed] [Google Scholar]
- 81. Díaz de León-Martínez L, Flores-Ramírez R, López-Mendoza CM, et al. Identification of volatile organic compounds in the urine of patients with cervical cancer. Test concept for timely screening. Clin Chim Acta 2021;522:132–140. [DOI] [PubMed] [Google Scholar]
- 82. V A B, Subramoniam M, Mathew L. Noninvasive detection of COPD and lung cancer through breath analysis using MOS Sensor array based e-nose. Expert Rev Mol Diagn 2021;21:1223–1233. [DOI] [PubMed] [Google Scholar]
- 83. Bannaga AS, Tyagi H, Daulton E, et al. Exploratory study using urinary volatile organic compounds for the detection of hepatocellular carcinoma. Molecules 2021;26:2447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Pinto J, Amaro F, Lima AR, et al. Urinary volatilomics unveils a candidate biomarker panel for noninvasive detection of clear cell renal cell carcinoma. J Proteome Res 2021;20:3068–3077. [DOI] [PubMed] [Google Scholar]
- 85. Long Y, Wang C, Wang T, et al. High performance exhaled breath biomarkers for diagnosis of lung cancer and potential biomarkers for classification of lung cancer. J Breath Res 2021;15:016017. [DOI] [PubMed] [Google Scholar]
- 86. Chen K, Liu L, Nie B, et al. Recognizing lung cancer and stages using a self-developed electronic nose system. Comput Biol Med 2021;131:104294. [DOI] [PubMed] [Google Scholar]
- 87. Mohamed N, van de Goor R, El-Sheikh M, et al. Feasibility of a portable electronic nose for detection of oral squamous cell carcinoma in Sudan. Healthcare (Basel) 2021;9:534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Boulind CE, Gould O, de Lacy Costello B, et al. Urinary volatile organic compound testing in fast-track patients with suspected colorectal cancer. Cancers (Basel) 2022;14:2127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Cheng HR, van Vorstenbosch RWR, Pachen DM, et al. Detecting colorectal adenomas and cancer using volatile organic compounds in exhaled breath: a proof-of-principle study to improve screening. Clin Transl Gastroenterol 2022 2022;13:e00518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Gasparri R, Capuano R, Guaglio A, et al. Volatolomic urinary profile analysis for diagnosis of the early stage of lung cancer. J Breath Res 2022;16:046008. [DOI] [PubMed] [Google Scholar]
- 91. Scheepers MHMC, Al-Difaie ZJJ, Wintjens AGWE, et al. Detection of differentiated thyroid carcinoma in exhaled breath with an electronic nose. J Breath Res 2022;16:036008. [DOI] [PubMed] [Google Scholar]
- 92. Sukaram T, Tansawat R, Apiparakoon T, et al. Exhaled volatile organic compounds for diagnosis of hepatocellular carcinoma. Sci Rep 2022;12:5326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Gashimova E, Temerdashev A, Porkhanov V, et al. Non-invasive exhaled breath and skin analysis to diagnose lung cancer: study of age effect on diagnostic accuracy. ACS Omega 2022;7:42613–42628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Anzivino R, Sciancalepore PI, Dragonieri S, et al. The role of a polymer-based e-Nose in the detection of head and neck cancer from exhaled breath. Sensors (Basel) 2022;22:6485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Liu Q, Fan Y, Zeng S, et al. Volatile organic compounds for early detection of prostate cancer from urine. Heliyon 2023;9:e16686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Costantini M, Filianoti A, Anceschi U, et al. Human urinary volatilome analysis in renal cancer by electronic nose. Biosensors (Basel) 2023;13:427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Li X, Wen X, Luo Z, et al. Development of a headspace-solid phase microextraction gas chromatography-high resolution mass spectrometry method for analyzing volatile organic compounds in urine: application in breast cancer biomarker discovery. Clin Chim Acta 2023;540:117236. [DOI] [PubMed] [Google Scholar]
- 98. Nakayama Y, Hanada M, Koda H, et al. Breast cancer detection using volatile compound profiles in exhaled breath via selected ion-flow tube mass spectrometry. J Breath Res 2022;17:016006. [DOI] [PubMed] [Google Scholar]
- 99. Liu J, Chen H, Li Y, et al. A novel non-invasive exhaled breath biopsy for the diagnosis and screening of breast cancer. J Hematol Oncol 2023;16:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Temerdashev AZ, Gashimova EM, Porkhanov VA, et al. Non-invasive lung cancer diagnostics through metabolites in exhaled breath: influence of the disease variability and comorbidities. Metabolites 2023;13:203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Sani SN, Zhou W, Ismail BB, et al. LC-MS/MS based volatile organic compound biomarkers analysis for early detection of lung cancer. Cancers (Basel) 2023;15:1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Sukaram T, Apiparakoon T, Tiyarattanachai T, et al. VOCs from exhaled breath for the diagnosis of hepatocellular carcinoma. Diagnostics (Basel) 2023;13:257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Kort S, Brusse-Keizer M, Schouwink H, et al. Diagnosing non-small cell lung cancer by exhaled breath profiling using an electronic nose: a multicenter validation study. Chest 2023;163:697–706. [DOI] [PubMed] [Google Scholar]
- 104. Cheng Y, Xie G, Chen T, et al. Distinct urinary metabolic profile of human colorectal cancer. J Proteome Res 2012;11:1354–1363. [DOI] [PubMed] [Google Scholar]
- 105. Silva CL, Passos M, Câmara JS. Investigation of urinary volatile organic metabolites as potential cancer biomarkers by solid-phase microextraction in combination with gas chromatography-mass spectrometry. Br J Cancer 2011;105:1894–1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Porto-Figueira P, Pereira JAM, Câmara JS. Exploring the potential of needle trap microextraction combined with chromatographic and statistical data to discriminate different types of cancer based on urinary volatomic biosignature. Anal Chim Acta 2018;1023:53–63. [DOI] [PubMed] [Google Scholar]
- 107. Qiu Y, Cai G, Su M, et al. Urinary metabonomic study on colorectal cancer. J Proteome Res 2010;9:1627–1634. [DOI] [PubMed] [Google Scholar]
- 108. Ning W, Qiao N, Zhang X, et al. Metabolic profiling analysis for clinical urine of colorectal cancer. Asia Pac J Clin Oncol 2021;17:403–413. [DOI] [PubMed] [Google Scholar]
- 109. Rozhentsov AA, Koptina AV, Mitrakov AA, et al. A new method to diagnose cancer based on image analysis of mass chromatograms of volatile organic compounds in urine. Sovremennye Tehnologii 2015;6:151–157. [Google Scholar]
- 110. Zhang Y, Guo L, Qiu Z, et al. Early diagnosis of breast cancer from exhaled breath by gas chromatography-mass spectrometry (GC/MS) analysis: a prospective cohort study. J Clin Lab Anal 2020;34:e23526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Kure S, Satoi S, Kitayama T, et al. A prediction model using 2-propanol and 2-butanone in urine distinguishes breast cancer. Sci Rep 2021;11:19801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Silva C, Perestrelo R, Silva P, et al. Volatomic pattern of breast cancer and cancer-free tissues as a powerful strategy to identify potential biomarkers. Analyst 2019;144:4153–4161. [DOI] [PubMed] [Google Scholar]
- 113. Wang C, Sun B, Guo L, et al. Volatile organic metabolites identify patients with breast cancer, cyclomastopathy, and mammary gland fibroma. Sci Rep 2014;4:5383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Li J, Peng Y, Liu Y, et al. Investigation of potential breath biomarkers for the early diagnosis of breast cancer using gas chromatography-mass spectrometry. Clin Chim Acta 2014;436:59–67. [DOI] [PubMed] [Google Scholar]
- 115. Mangler M, Freitag C, Lanowska M, et al. Volatile organic compounds (VOCs) in exhaled breath of patients with breast cancer in a clinical setting. Ginekol Pol 2012;83:730–736. [PubMed] [Google Scholar]
- 116. Taunk K, Taware R, More TH, et al. A non-invasive approach to explore the discriminatory potential of the urinary volatilome of invasive ductal carcinoma of the breast. RSC Adv 2018;8:25040–25050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Jaeschke R, Guyatt GH, Sackett DL. Users’ guides to the medical literature. III. How to use an article about a diagnostic test. B. What are the results and will they help me in caring for my patients? The Evidence-Based Medicine Working Group. JAMA 1994;271:703–707. [DOI] [PubMed] [Google Scholar]
- 118. Fagan TJ. Letter: Nomogram for Bayes theorem. N Engl J Med 1975;293:257. [DOI] [PubMed] [Google Scholar]
- 119. van der Sar IG, Wijbenga N, Nakshbandi G, et al. The smell of lung disease: a review of the current status of electronic nose technology. Respir Res 2021;22:246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016;16:341–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Zhao L, Bin S, He HL, et al. Sodium butyrate increases P-gp expression in lung cancer by upregulation of STAT3 and mRNA stabilization of ABCB1. Anticancer Drugs 2018;29:227–233. [DOI] [PubMed] [Google Scholar]
- 122. Zhang Y, Zhou L, Bao YL, et al. Butyrate induces cell apoptosis through activation of JNK MAP kinase pathway in human colon cancer RKO cells. Chem Biol Interact 2010;185:174–181. [DOI] [PubMed] [Google Scholar]
- 123. Salimi V, Shahsavari Z, Safizadeh B, et al. Sodium butyrate promotes apoptosis in breast cancer cells through reactive oxygen species (ROS) formation and mitochondrial impairment. Lipids Health Dis 2017;16:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Singh V, Yeoh BS, Chassaing B, et al. Dysregulated microbial fermentation of soluble fiber induces cholestatic liver cancer. Cell 2018;175:679–694.e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The main data supporting the findings of this study are included within the article and its supplementary materials. Additional data are available from the corresponding authors upon reasonable request.









