Minireviews Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Methodol. Mar 26, 2017; 7(1): 9-15
Published online Mar 26, 2017. doi: 10.5662/wjm.v7.i1.9
Chromogranin A as a valid marker in oncology: Clinical application or false hopes?
Stavros Gkolfinopoulos, Demetris Papamichael, Panteleimon Kountourakis, Department of Medical Oncology, BOC Oncology Center, 2006 Strovolos, Nicosia, Cyprus
Konstantinos Tsapakidis, Department of Medical Oncology, University Hospital of Larissa, 41110 Larissa, Greece
Konstantinos Papadimitriou, Department of Medical Oncology, University Hospital of Antwerp, 2610 Antwerp, Belgium
Author contributions: Gkolfinopoulos S performed the majority of the manuscript preparation; Tsapakidis K contributed to the manuscript preparation; Papadimitriou K contributed to the manuscript preparation; Papamichael D contributed to the manuscript preparation and revision; Kountourakis P contributed to conception, designed the study, coordinated manuscript preparation, prepared the revised version.
Conflict-of-interest statement: There is no conflict of interest on the part of any other authors on this study.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Panteleimon Kountourakis, MD, PhD, Department of Medical Oncology, BOC Oncology Center, 32 Acropoleos Ave, 2006 Strovolos, Nicosia, Cyprus. pantkount@gmail.com
Telephone: +357-22-847402 Fax: +357-22-511870
Received: October 16, 2016
Peer-review started: October 19, 2016
First decision: November 30, 2016
Revised: December 15, 2016
Accepted: January 2, 2017
Article in press: January 4, 2017
Published online: March 26, 2017

Abstract

Chromogranin A, due to its primary expression throughout the neuroendocrine system, is a widely accepted biomarker for the assessment of neuro-endocrine tumors. It has been traditionally used in the management of patients with tumors of gastro-enteropancreatic origin. Lately, it has also been implicated in various conditions and diseases, both benign and malignant. However, the paucity of data of adequate strength, as well as its relation with common physiologic conditions and its interaction with commonly prescribed medications, limit its clinical use in only a narrow spectrum. Herein, we present a thorough review to the most frequent conditions where its levels are affected, focusing specifically on its potential use as a prognostic and predictive biomarker in oncology.

Key Words: Cancer, Neuroendocrine tumors, Prognosis, Chromogranin A, Biomarker

Core tip: In the era of targeted therapy, there is an unmet need for the development of more sensitive, specific and reliable biomarkers for early diagnosis, prognosis and detection of early recurrence to tumors which comprise an extremely heterogeneous group.



INTRODUCTION

The Granins comprise a family of proteins whose most well known members are chromogranin A (CgA), chromogranin B (CgB) and secretogranin II, with their most common characteristic being their acidic profile. They are produced as pre-proteins in the ribosomes and subsequently they undergo post-translational modifications in the endoplasmic reticulum and in the Golgi apparatus[1]. It has been shown that they are co-stored with peptides and amines in the granules of endocrine cells. They can also be found in a number of other cells, including immune cells, epithelial cells and peripheral neurons[2]. Other proteins that are also included in the granin family are secretogranin III, the HISL-19 antigen (secretogranin IV), the neuroendocrine secretory protein 7B2 (secretogranin V), NESP55 (secretogranin VI) and nerve growth factor-inducible protein VGF (secretogranin VII)[3].

Granins are composed of single-polypeptide chains of approximately 180 to 700 amino acids, with CgA being a 49 kDa protein produced mainly by endocrine and neuroendocrine cells[1,4,5]. It was first discovered in the chromaffin granules of the adrenal medulla, where it is stored along with the resident hormones, like calcitonin, and then secreted with them[5]. The CgA gene, located on chromosome 14, is probably a single copy gene rather than a member of a dispersed, multigene family[6].

Since the discovery of CgA and its pathologically high levels in patients with neuroendocrine tumors, it has been correlated with a number of other conditions, both benign and malignant (Tables 1 and 2). Its sensitivity and specificity in each one of these conditions differ significantly, depending on various factors, limiting its use as an effective prognostic and/or predictive marker in a narrow spectrum of conditions. This review summarizes the most frequent conditions where CgA levels are affected, focusing specifically on its function as a biomarker in oncology.

Table 1 Non cancerous causes of chromogranin A elevation.
Disease
CardiovascularEndocrineGastrointestinalInflammatory
Acute coronary syndromeHyperparathyroidismChronic atrophic gastritisChronic bronchitis
Arterial hypertensionHyperthyroidismChronic hepatitisChronic obstructive pulmonary disease
Cardiac insufficiencyHypercortisolismInflammatory/irritable bowel syndromeGiant cell arthritis
Liver cirrhosisRheumatoid arthritis
PancreatitisSystemic inflammatory response syndrome
Drugs
CorticoidsH2 receptor antagonistProton pump inhibitor
Status
ExerciseIngestion of a mealPregnancy
Factors having potential influence on sample
Fibrin presenceHaemolysisImposing effect: Autoantibodies presence (RF-IgM, Avidine, Heterofile)Late afternoon/night > morning
LipaemiaPlasma > serum-
Table 2 Frequent cancer-related causes of increased chromogranin A.
CancerNeuroendocrine tumors
BreastColorectal
ColonGastric
HepatocellularMedullary thyroid
OvarianNeuroblastoma
PancreaticPancreatic
ProstateParaganglioma
Pheochromocytoma
Pituitary
Small cell lung
Small intestinal

CgA may be secreted in the blood in its full length or in fragments after cleavage. These fragment peptides include Catestin, Chromacin, Pancreastatin, Parastatin, Vasostatin I, Vasostatin II and WE-14[1]. Although CgA and its peptides definite functions have not been fully understood, it is believed that they are important factors for the formation and regulation of dense-core granules, heart function, catecholamines and parathyroid hormone secretion, carbohydrate and lipid metabolism, immune properties and reproduction[7].

CGA IN NON-MALIGNANT DISEASES AND CONDITIONS

CgA has been correlated with a wide range of non-malignant systemic diseases, including hypertension, heart and hepatic failure (Table 1)[1,8]. It is produced by the human myocardium and exerts negative inotropic effect, so in chronic heart failure it is significantly elevated and its levels can parallel the severity of cardiac dysfunction and could be used as an independent predictor of mortality[8]. Furthermore, basal plasma CgA levels correlate with sympathetic tone and increased adrenal sympathetic nerve activity. Subsequently, CgA levels are usually elevated in hypertension[8].

Furthermore, it can be raised in renal insufficiency, as a result of decreased plasma clearance. It has also been implicated in inflammatory and autoimmune conditions, like Rheumatoid arthritis[9,10]. Furthermore, PPIs, which are some of the most commonly prescribed drugs, may cause a secondary increase in CgA levels due to increased gastrin production[11]. Another common condition that is associated with elevated levels of CgA, is chronic atrophic gastritis (Table 1)[12]. Summarizing, in non malignant diseases and conditions, CgA values may reach values of hundreds (ng/mL), but it is very uncommon to reach levels of several thousands that could be consistent with cancer diagnosis.

CGA IN MALIGNANT DISEASES
Bronchopulmonary neuroendocrine tumors

In small cell lung carcinomas (SCLC) the mean CgA plasma levels are higher than those found in normal controls or in patients with chronic obstructive pulmonary disease, lung adenocarcinoma and large-cell lung carcinoma. The levels of CgA are associated with the extent of the disease, but the levels of NSE have been proven to be more accurate in that regard[13-16]. Bronchopulmonary neuroendocrine tumors (BP-NETs) comprise approximately 20% of all lung cancers and represent a spectrum of tumors arising from neuroendocrine cells of the BP-epithelium. Although they share structural, morphological, immunohistochemical, and ultrastructural features, they are separated into 4 subgroups: Typical carcinoid tumour (TC), atypical carcinoid tumour (AC), large-cell neuroendocrine carcinoma (LC-NEC), and SCLC[17]. The diagnosis is based on the recognition of neuroendocrine morphology, such as organoid pattern, and on the immunohistochemical demonstration of specific neuroendocrine markers, like chromogranin, synaptophysin, and neural cell adhesion molecule (NCAM), also known as CD 56. To confirm the neuroendocrine origin of the tumour cells, at least one of those markers must be positive[18]. Although they can produce a variety of peptides and hormones, like gastrin-releasing peptide (bombesin) and 5-hydroxytryptophan, bronchial NETs only occasionally secrete bioactive products that can easily be measured. As a result, elevated plasma or urinary hormone levels are only rarely detected. Serum levels of CgA are lower in bronchial NETs than those observed in NETs of other sites, and they overlap with those seen in patients who have non-malignant conditions associated with increased CgA levels[17].

Breast cancer

In breast cancer CgA was discovered both in epithelial cells of normal mammary gland as well as in breast cancer. However, it does not seem to offer any additional information about the presence, the extent and the histology of breast cancer when compared to the more established Ca 15-3. Furthermore, serum CgA was not sensitive enough to identify the rarely encountered subtype of breast cancer with neuroendocrine differentiation[19].

Merkel cell carcinoma

Merkel cell carcinoma (MCC) is a rare, aggressive, cutaneous malignancy with neuroendocrine differentiation, that predominantly affects older adults with light skin complexion. MCC has a propensity for local recurrence and regional lymph node metastases. On immunohistochemistry, the tumour cells show features of both epithelial and neuroendocrine origin, including the expression of CgA. CgA blood levels are used by many physicians as a predictive marker for the response of the tumour to chemotherapy, though it has never been shown to correlate with progression-free survival, disease specific survival, or disease recurrence[20].

Gastroenteropancreatic neuroendocrine tumors

Chromogranins were early discovered to be elevated in the plasma of patients with neuroendocrine tumors[21,22]. They arise from neuroendocrine cells that occur throughout the length of the entire gut, and about two-thirds of them are of gastrointestinal or pancreatic origin (GEP-NETs)[23]. Their relevance in the diagnosis, prognosis, clinical evaluation after cytoreductive surgery, and subsequent follow-up of patients with those types of tumors, has been studied for more than 20 years[21].

Although GEP-NETs excrete a number of peptides specific to the neuroendocrine cell of origin, CgA is the most frequently studied biomarker for their diagnosis and subsequent follow-up[24-26]. Not all GEP-NETs produce CgA, but for those that do, elevated circulating levels of CgA could be related with tumour burden as well as recurrence, and are considered a marker of poor prognosis and reduced survival in both ileal and pancreatic NETs[27,28]. For example, in patients with midgut carcinoids the 5-year OS was estimated to be 22% with CgA levels > 75 nmol/L, while it was raised to 63% with levels lower than this value. The decrease of CgA levels has also been used as a marker of response to treatment in clinical trials, where biochemical response is defined as a ≥ 50% reduction of CgA[29].

The highest levels of CgA are observed in patients with functioning ileal NET and carcinoid syndrome, followed by those with liver metastases. Metastatic disease in the lymph nodes does not seem to cause a significant increase in the levels of CgA[27,29]. However, its value in predicting liver metastases, as compared to morphological tumour changes as measured by CT or MRI, is limited, with a sensitivity and specificity of 71% and 50% respectively[30]. On the contrary, it should be noted that its elevation, even in values of several thousands (ng/mL), could be not related with deterioration of clinical status.

The overall sensitivity of CgA in the diagnosis of neuroendocrine tumors is around 60%-80% and depends on the primary site, on the degree of differentiation and on the status of the disease[31]. This marker has a low sensitivity regarding its use in distinguishing the different types of NETs. It should be noted also, that the specificity and sensitivity of the assay for CgA measurement differ between the available commercial kits[32].

Moreover, the use of CgA as a diagnostic biomarker in GEP-NETs has certain limitations. Firstly, although CgA could be useful in predicting tumor relapse or progression, with rapidly increasing levels correlating with shorter survival, it should be noted that CgA levels are also affected by the secretory activity of a functioning tumor. This has particular importance in patients treated with somatostatin analogues (SSAs), where the drop in CgA levels may reflect the inhibition of the secretory activity of the tumour rather than a true anti-tumour effect[33].

Midgut carcinoids have often been misdiagnosed as irritable bowel syndrome or inflammatory bowel disease, where CgA may also be increased, due to the common manifestation of watery diarrheas[34].

CgA along with NSE have been retrospectively studied as prognostic biomarkers in GEP-NETs[35]. In a phase II study of Everolimus in GEP-NETs it has been demonstrated that higher baseline levels of CgA were associated with shorter PFS, while the patients with the shortest PFS had elevated concentrations of both CgA and NSE at baseline. In that same study, CgA and NSE responses were defined as a 50% or greater reduction from baseline or normalization, and early CgA and NSE responses were defined as a 30% or greater decrease from baseline or normalization after 4 wk of treatment. For both those markers, an early decrease predicted for clinical benefit, which, in the case of CgA, meant both longer PFS (13.3 mo vs 7.5 mo; HR = 0.25; P < 0.001) and longer OS (24.9 mo vs 12.7 mo; HR = 0.4; P = 0.01)[36].

Those results have been confirmed in a relevant analysis of the phase III RADIANT-2 clinical trial, where it was shown that early decrease of CgA levels by Everolimus can be used as a surrogate marker of PFS in this setting[37]. To our knowledge, no such data exist for patients with GEP-NETs treated with Sunitinib.

There is no doubt that due to the existing data, CgA role in NET diagnosis is strongly limited and debated. Therefore, it could not be recommended and applied in our daily clinical practice. Moreover, it could be used primarily but with caution, in NETs as a marker of therapy response.

Prostate cancer

CgA is excreted by the neuroendocrine cells that are dispersed throughout the prostatic gland. Neuroendocrine cells can be found in the normal prostate as well as in benign prostate hyperplasia and in primary or metastatic prostatic adenocarcinoma[38]. In addition to CgA, neuroendocrine cells produce a variety of biogenic amines, such as NSE, calcitonin and somatostatin. According to their degree of differentiation, prostatic malignant neuroendocrine cells may continue to produce those amines, though they differ in their morphology from their normal counterparts[39].

Although not specific for prostate cancer, there is evidence that high levels of serum CgA are a marker of advanced disease, associated both with high tumor grade and later stage[40]. High levels also characterize the shift from a disease responding to androgen deprivation therapies (ADT) to an androgen-independent, aggressive malignancy[41,42]. Pathophysiologically, this is to be expected, since an increase in circulating CgA and NSE reflect tissue neuroendocrine differentiation. There is evidence that the degree of neuroendocrine differentiation increases with prostate cancer progression, and it has been suggested that it constitutes a major mechanism of resistance to ADT[38]. Neuroendocrine cells do not express androgen receptors, consequently they are not regulated by androgens[43].

There is also evidence that serum CgA, either alone or combined with serum PSA, may predict poor prognosis in castration-resistant prostate cancer following endocrine therapy[44-46]. Moreover, circulating neuroendocrine peptides have been linked with angiogenesis and invasive potential[39,47]. However, serum concentration of CgA and tissue IHC expression do not show robust correlation and CgA does not seem to positively correlate with treatment response to cytotoxic chemotherapy in metastatic prostate cancer with neuroendocrine differentiation[48].

Multiple Endocrine Neoplasia type 1 syndrome

Multiple Endocrine Neoplasia type 1 (MEN 1) is a rare hereditary autosomal dominant endocrine cancer syndrome, that is characterized by the development of tumors, both benign and malignant, in multiple endocrine organs. The tumors most often appear in the parathyroid glands, in the endocrine cells dispersed throughout the gastroenteropancreatic (GEP) tract and in the anterior pituitary, though other endocrine and non-endocrine tumors have also been reported, namely adrenocortical and thyroid tumors, visceral and cutaneous lipomas, meningiomas, facial angiofibromas and collagenomas, and thymic, gastric, and bronchial carcinoids[49].

Several studies have assessed the role of CgA in demonstrating the presence of a GEP-NET in MEN 1 syndrome. It has been confirmed that abnormally elevated CgA levels are highly suggestive of both sporadic and MEN 1-related GEP-NETs. The highest levels are observed in metastatic disease, especially when the metastases are located in the liver, and in functioning tumors, especially in gastrinomas[50]. In MEN 1 patients without biochemical or imaging evidence of GEP tumors, the data are scanty and conflicting. Some studies have reported increased CgA levels in 11%-33% of patients with pituitary adenomas, both secreting and non-functioning[51]. In addition, conflicting data have been published regarding the relationship between CgA levels and hyperparathyroidism, either primary or in the context of MEN 1 syndrome[52]. However, it appears that the generalised hyperplasia of the endocrine system, that occurs in MEN 1 syndrome, tends to lead to at least mildly elevated levels of circulating CgA, while markedly raised levels may indicate the presence of a GEP-NET[50].

Hepatocellular carcinoma

Hepatocellular carcinoma (HCC) represents the most frequent complication and a major cause of death in patients with cirrhosis of any aetiology[53]. The most widely used biomarker for diagnosis and follow-up is AFP[54]. CgA has been found elevated in patients with liver cirrhosis and in those with HCC[55,56]. However, its use as a diagnostic biomarker for the presence of HCC in the context of cirrhosis should be discouraged, since the levels of CgA have not been found to differ significantly between these two conditions[54]. The prognostic meaning of CgA in HCC has yet to be elucidated.

DISCUSSION

The extent of the physiological functions of CgA indicates its potential role as a biomarker in a wide spectrum of benign and malignant diseases (Tables 1 and 2). However, certain factors limit its usefulness in only a few. There is a lack of prospective studies that aim to evaluate its validity in the diagnosis and prognosis of specific conditions.

Although limitations exist, CgA is the most studied biomarker for GEP-NETs’ diagnosis and management. Clinicians should be aware of the variation of measurements by numerous physiologic and pathologic conditions, its limited predictive value and the modest sensitivity (Table 3)[57-62]. Moreover, data support that baseline CgA levels and changes during treatment are prognostic. Even, its specificity could be heavily affected by several benign conditions, also intrinsic features of the disease could be related with the high variability of CgA values[63]. Diagnostic accuracy of CgA for GEP-NETs appear to be higher for well vs poorly differentiated tumors, functioning vs non-functioning, metastatic vs locoregional disease. There is no doubt that it is more reliable when used to evaluate response to therapy or disease progression than early diagnosis or recurrence.

Table 3 Chromogranin A diagnostic accuracy in neuroendocrine tumor studies.
Type (no pts)CgA cut-offSensitivity (%)Specificity (%)Ref.
NET (128)100 μg/L5968[57]
NET (127)34.7 u/L67.985.7[35]
NET (80)17 u/L56.3100[58]
NET (63)34 u/L5594[59]
GEP/NET (61)20 u/L9283[50]
100 u/L4799
GEP/NET (124)130 μg/L62.998.4[16]
GEP/NET (202)53 ng/mL71.377.8[60]
NET (120)98 ng/mL79NA[61]
GEP/NET (119)2.8 nmol/L92.9100[62]

It should be underlined that there are many assays and commercial kits available for CgA levels evaluation, thus very strict quality assurance and standardization should be used. In addition, CgA evaluation is more convenient than U5-HIAA, which requires a 24-h urine collection and 3 d before the collection a dietary abstinence from tryptophan/serotonin-rich foods.

Finally, in cancers where a biomarker is already in use, such as AFP in hepatocellular carcinoma or Ca 15-3 in breast cancer, CgA has not been proven to be of greater diagnostic and/or prognostic value than the currently used biomarker. It also provides an indication for the presence of a strong component of neuroendocrine differentiation within an adenocarcinoma. That also applies to cases of prostatic adenocarcinoma that develop resistance to androgen deprivation therapy during the progression of the disease, as a result of the gradual shift of the tumor cells towards a neuroendocrine phenotype. The early recognition of that phenomenon may lead to an earlier change in the treatment strategy, which, in turn, may prove to provide clinical benefit. Moreover, it should be used with caution and only in comparison with other methods of determining the course of the disease, such as radiologic and histological evaluation, simply because there are not enough data to support its use as a single, stand-alone marker.

CONCLUSION

Due to the fact that NET symptoms could be vague, or even the disease course may be asymptomatic, diagnosis could be delayed for many years. There is an unmet need for the development of more sensitive, specific and reliable biomarkers for early diagnosis, prognosis and detection of early recurrence to these tumors which comprise an extremely heterogeneous group. Multianalyte assays focusing on novel analytes, such as microRNA, gene transcripts, and circulating tumor cells could be an interesting area for further research given the fact that is unlikely any single marker to be effective.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Clinical medicine

Country of origin: Cyprus

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): D

Grade E (Poor): 0

P- Reviewer: Deutsch JC, Ke YQ, Ni Y, Ooi LLPJ S- Editor: Ji FF L- Editor: A E- Editor: Lu YJ

References
1.  Taupenot L, Harper KL, O’Connor DT. The chromogranin-secretogranin family. N Engl J Med. 2003;348:1134-1149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 632]  [Cited by in F6Publishing: 604]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
2.  Eriksson B, Arnberg H, Oberg K, Hellman U, Lundqvist G, Wernstedt C, Wilander E. A polyclonal antiserum against chromogranin A and B--a new sensitive marker for neuroendocrine tumours. Acta Endocrinol (Copenh). 1990;122:145-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
3.  Helle KB. Chromogranins A and B and secretogranin II as prohormones for regulatory peptides from the diffuse neuroendocrine system. Results Probl Cell Differ. 2010;50:21-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 15]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
4.  Campana D, Nori F, Piscitelli L, Morselli-Labate AM, Pezzilli R, Corinaldesi R, Tomassetti P. Chromogranin A: is it a useful marker of neuroendocrine tumors? J Clin Oncol. 2007;25:1967-1973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 179]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
5.  Deftos LJ. Chromogranin A: its role in endocrine function and as an endocrine and neuroendocrine tumor marker. Endocr Rev. 1991;12:181-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 165]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
6.  Murray SS, Deaven LL, Burton DW, O’Connor DI, Mellon PL, Deftos LJ. The gene for human chromogranin A (CgA) is located on chromosome 14. Biochem Biophys Res Commun. 1987;142:141-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
7.  Lawrence B, Gustafsson BI, Kidd M, Pavel M, Svejda B, Modlin IM. The clinical relevance of chromogranin A as a biomarker for gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am. 2011;40:111-134, viii.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 111]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
8.  Goetze JP, Alehagen U, Flyvbjerg A, Rehfeld JF. Chromogranin A as a biomarker in cardiovascular disease. Biomark Med. 2014;8:133-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
9.  di Comite G, Marinosci A, Di Matteo P, Manfredi A, Rovere-Querini P, Baldissera E, Aiello P, Corti A, Sabbadini MG. Neuroendocrine modulation induced by selective blockade of TNF-alpha in rheumatoid arthritis. Ann N Y Acad Sci. 2006;1069:428-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
10.  Di Comite G, Rossi CM, Marinosci A, Lolmede K, Baldissera E, Aiello P, Mueller RB, Herrmann M, Voll RE, Rovere-Querini P. Circulating chromogranin A reveals extra-articular involvement in patients with rheumatoid arthritis and curbs TNF-alpha-elicited endothelial activation. J Leukoc Biol. 2009;85:81-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 47]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
11.  Sanduleanu S, Stridsberg M, Jonkers D, Hameeteman W, Biemond I, Lundqvist G, Lamers C, Stockbrügger RW. Serum gastrin and chromogranin A during medium- and long-term acid suppressive therapy: a case-control study. Aliment Pharmacol Ther. 1999;13:145-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 85]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
12.  Glinicki P, Jeske W. Chromogranin A (CgA)--the influence of various factors in vivo and in vitro, and existing disorders on it’s concentration in blood. Endokrynol Pol. 2010;61:384-387.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Modlin IM, Gustafsson BI, Moss SF, Pavel M, Tsolakis AV, Kidd M. Chromogranin A--biological function and clinical utility in neuro endocrine tumor disease. Ann Surg Oncol. 2010;17:2427-2443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 244]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
14.  Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, Thakker RV, Caplin M, Delle Fave G, Kaltsas GA, Krenning EP. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol. 2008;9:61-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1131]  [Cited by in F6Publishing: 1119]  [Article Influence: 69.9]  [Reference Citation Analysis (0)]
15.  Nobels FR, Kwekkeboom DJ, Coopmans W, Schoenmakers CH, Lindemans J, De Herder WW, Krenning EP, Bouillon R, Lamberts SW. Chromogranin A as serum marker for neuroendocrine neoplasia: comparison with neuron-specific enolase and the alpha-subunit of glycoprotein hormones. J Clin Endocrinol Metab. 1997;82:2622-2628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 73]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
16.  Nehar D, Lombard-Bohas C, Olivieri S, Claustrat B, Chayvialle JA, Penes MC, Sassolas G, Borson-Chazot F. Interest of Chromogranin A for diagnosis and follow-up of endocrine tumours. Clin Endocrinol (Oxf). 2004;60:644-652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 131]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
17.  Gustafsson BI, Kidd M, Chan A, Malfertheiner MV, Modlin IM. Bronchopulmonary neuroendocrine tumors. Cancer. 2008;113:5-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 347]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
18.  Yuan C, Keating B, Farricielli LA, Zhang K. Large-cell neuroendocrine carcinoma (LCNEC) without pulmonary symptoms diagnosed in a cutaneous metastasis. Am J Case Rep. 2014;15:97-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
19.  Giovanella L, Marelli M, Ceriani L, Giardina G, Garancini S, Colombo L. Evaluation of chromogranin A expression in serum and tissues of breast cancer patients. Int J Biol Markers. 2001;16:268-272.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Gaiser MR, Daily K, Hoffmann J, Brune M, Enk A, Brownell I. Evaluating blood levels of neuron specific enolase, chromogranin A, and circulating tumor cells as Merkel cell carcinoma biomarkers. Oncotarget. 2015;6:26472-26482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
21.  O’Connor DT, Deftos LJ. Secretion of chromogranin A by peptide-producing endocrine neoplasms. N Engl J Med. 1986;314:1145-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in F6Publishing: 363]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
22.  Eriksson B, Arnberg H, Oberg K, Hellman U, Lundqvist G, Wernstedt C, Wilander E. Chromogranins--new sensitive markers for neuroendocrine tumors. Acta Oncol. 1989;28:325-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
23.  Verbeek WH, Korse CM, Tesselaar ME. GEP-NETs UPDATE: Secreting gastro-enteropancreatic neuroendocrine tumours and biomarkers. Eur J Endocrinol. 2016;174:R1-R7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
24.  Kanakis G, Kaltsas G. Biochemical markers for gastroenteropancreatic neuroendocrine tumours (GEP-NETs). Best Pract Res Clin Gastroenterol. 2012;26:791-802.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
25.  Arnold R, Chen YJ, Costa F, Falconi M, Gross D, Grossman AB, Hyrdel R, Kos-Kudła B, Salazar R, Plöckinger U. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: follow-up and documentation. Neuroendocrinology. 2009;90:227-233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 88]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
26.  Oberg K. Circulating biomarkers in gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer. 2011;18 Suppl 1:S17-S25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
27.  Arnold R, Wilke A, Rinke A, Mayer C, Kann PH, Klose KJ, Scherag A, Hahmann M, Müller HH, Barth P. Plasma chromogranin A as marker for survival in patients with metastatic endocrine gastroenteropancreatic tumors. Clin Gastroenterol Hepatol. 2008;6:820-827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 130]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
28.  Welin S, Stridsberg M, Cunningham J, Granberg D, Skogseid B, Oberg K, Eriksson B, Janson ET. Elevated plasma chromogranin A is the first indication of recurrence in radically operated midgut carcinoid tumors. Neuroendocrinology. 2009;89:302-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 80]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
29.  Ardill JE, Erikkson B. The importance of the measurement of circulating markers in patients with neuroendocrine tumours of the pancreas and gut. Endocr Relat Cancer. 2003;10:459-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 59]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
30.  Vezzosi D, Walter T, Laplanche A, Raoul JL, Dromain C, Ruszniewski P, d’Herbomez M, Guigay J, Mitry E, Cadiot G. Chromogranin A measurement in metastatic well-differentiated gastroenteropancreatic neuroendocrine carcinoma: screening for false positives and a prospective follow-up study. Int J Biol Markers. 2011;26:94-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
31.  Korse CM, Taal BG, Vincent A, van Velthuysen ML, Baas P, Buning-Kager JC, Linders TC, Bonfrer JM. Choice of tumour markers in patients with neuroendocrine tumours is dependent on the histological grade. A marker study of Chromogranin A, Neuron specific enolase, Progastrin-releasing peptide and cytokeratin fragments. Eur J Cancer. 2012;48:662-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 72]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
32.  O’Connor DT, Pandlan MR, Carlton E, Cervenka JH, Hslao RJ. Rapid radioimmunoassay of circulating chromogranin A: in vitro stability, exploration of the neuroendocrine character of neoplasia, and assessment of the effects of organ failure. Clin Chem. 1989;35:1631-1637.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Baudin E, Bidart JM, Bachelot A, Ducreux M, Elias D, Ruffié P, Schlumberger M. Impact of chromogranin A measurement in the work-up of neuroendocrine tumors. Ann Oncol. 2001;12 Suppl 2:S79-S82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
34.  Sidhu R, McAlindon ME, Leeds JS, Skilling J, Sanders DS. The role of serum chromogranin A in diarrhoea predominant irritable bowel syndrome. J Gastrointestin Liver Dis. 2009;18:23-26.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Bajetta E, Ferrari L, Martinetti A, Celio L, Procopio G, Artale S, Zilembo N, Di Bartolomeo M, Seregni E, Bombardieri E. Chromogranin A, neuron specific enolase, carcinoembryonic antigen, and hydroxyindole acetic acid evaluation in patients with neuroendocrine tumors. Cancer. 1999;86:858-865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
36.  Yao JC, Pavel M, Phan AT, Kulke MH, Hoosen S, St Peter J, Cherfi A, Öberg KE. Chromogranin A and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with everolimus. J Clin Endocrinol Metab. 2011;96:3741-3749.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 162]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
37.  Baudin E, Wolin EM, Castellano D, Kaltsas G, Lebrec J, Tsuchihashi Z, Klimovsky J, Saletan S, Yao JC, Gross D. Effect of everolimus plus octreotide LAR treatment on chromogranin A and 5-hydroxyindoleacetic acid levels in patients with advanced neuroendocrine tumors: Phase III RADIANT-2 study results. J Clin Oncol. 2011;29 Suppl:Abstr 10527.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Parimi V, Goyal R, Poropatich K, Yang XJ. Neuroendocrine differentiation of prostate cancer: a review. Am J Clin Exp Urol. 2014;2:273-285.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Conteduca V, Aieta M, Amadori D, De Giorgi U. Neuroendocrine differentiation in prostate cancer: current and emerging therapy strategies. Crit Rev Oncol Hematol. 2014;92:11-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 63]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
40.  Bocan EV, Mederle O, Sârb S, Minciu R, Agapie D, Raica M. Correlation between histopathological form and the degree of neuroendocrine differentiations in prostate cancer. Rom J Morphol Embryol. 2011;52:1215-1218.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Hirano D, Okada Y, Minei S, Takimoto Y, Nemoto N. Neuroendocrine differentiation in hormone refractory prostate cancer following androgen deprivation therapy. Eur Urol. 2004;45:586-592; discussion 592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 221]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
42.  Berruti A, Mosca A, Porpiglia F, Bollito E, Tucci M, Vana F, Cracco C, Torta M, Russo L, Cappia S. Chromogranin A expression in patients with hormone naïve prostate cancer predicts the development of hormone refractory disease. J Urol. 2007;178:838-843; quiz 1129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 57]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
43.  Bonkhoff H, Stein U, Remberger K. Androgen receptor status in endocrine-paracrine cell types of the normal, hyperplastic, and neoplastic human prostate. Virchows Arch A Pathol Anat Histopathol. 1993;423:291-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 107]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
44.  Isshiki S, Akakura K, Komiya A, Suzuki H, Kamiya N, Ito H. Chromogranin a concentration as a serum marker to predict prognosis after endocrine therapy for prostate cancer. J Urol. 2002;167:512-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 59]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
45.  Berruti A, Dogliotti L, Mosca A, Gorzegno G, Bollito E, Mari M, Tarabuzzi R, Poggio M, Torta M, Fontana D. Potential clinical value of circulating chromogranin A in patients with prostate carcinoma. Ann Oncol. 2001;12 Suppl 2:S153-S157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
46.  Khan MO, Ather MH. Chromogranin A--serum marker for prostate cancer. J Pak Med Assoc. 2011;61:108-111.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Hoosein NM, Logothetis CJ, Chung LW. Differential effects of peptide hormones bombesin, vasoactive intestinal polypeptide and somatostatin analog RC-160 on the invasive capacity of human prostatic carcinoma cells. J Urol. 1993;149:1209-1213.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Berruti A, Mosca A, Tucci M, Terrone C, Torta M, Tarabuzzi R, Russo L, Cracco C, Bollito E, Scarpa RM. Independent prognostic role of circulating chromogranin A in prostate cancer patients with hormone-refractory disease. Endocr Relat Cancer. 2005;12:109-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 102]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
49.  Eriksson B, Oberg K, Skogseid B. Neuroendocrine pancreatic tumors. Clinical findings in a prospective study of 84 patients. Acta Oncol. 1989;28:373-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
50.  Peracchi M, Conte D, Gebbia C, Penati C, Pizzinelli S, Arosio M, Corbetta S, Spada A. Plasma chromogranin A in patients with sporadic gastro-entero-pancreatic neuroendocrine tumors or multiple endocrine neoplasia type 1. Eur J Endocrinol. 2003;148:39-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
51.  Deftos LJ, O’Connor DT, Wilson CB, Fitzgerald PA. Human pituitary tumors secrete chromogranin-A. J Clin Endocrinol Metab. 1989;68:869-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
52.  Nanes MS, O’Connor DT, Marx SJ. Plasma chromogranin-A in primary hyperparathyroidism. J Clin Endocrinol Metab. 1989;69:950-955.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 12]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
53.  Sangiovanni A, Del Ninno E, Fasani P, De Fazio C, Ronchi G, Romeo R, Morabito A, De Franchis R, Colombo M. Increased survival of cirrhotic patients with a hepatocellular carcinoma detected during surveillance. Gastroenterology. 2004;126:1005-1014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 437]  [Cited by in F6Publishing: 424]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
54.  Massironi S, Fraquelli M, Paggi S, Sangiovanni A, Conte D, Sciola V, Ciafardini C, Colombo M, Peracchi M. Chromogranin A levels in chronic liver disease and hepatocellular carcinoma. Dig Liver Dis. 2009;41:31-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 27]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
55.  Spadaro A, Ajello A, Morace C, Zirilli A, D’arrigo G, Luigiano C, Martino F, Bene A, Migliorato D, Turiano S. Serum chromogranin-A in hepatocellular carcinoma: diagnostic utility and limits. World J Gastroenterol. 2005;11:1987-1990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
56.  Leone N, Pellicano R, Brunello F, Rizzetto M, Ponzetto A. Elevated serum chromogranin A in patients with hepatocellular carcinoma. Clin Exp Med. 2002;2:119-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
57.  Baudin E, Gigliotti A, Ducreux M, Ropers J, Comoy E, Sabourin JC, Bidart JM, Cailleux AF, Bonacci R, Ruffié P. Neuron-specific enolase and chromogranin A as markers of neuroendocrine tumours. Br J Cancer. 1998;78:1102-1107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 174]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
58.  Tomassetti P, Migliori M, Simoni P, Casadei R, De Iasio R, Corinaldesi R, Gullo L. Diagnostic value of plasma chromogranin A in neuroendocrine tumours. Eur J Gastroenterol Hepatol. 2001;13:55-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 92]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
59.  Cimitan M, Buonadonna A, Cannizzaro R, Canzonieri V, Borsatti E, Ruffo R, De Apollonia L. Somatostatin receptor scintigraphy versus chromogranin A assay in the management of patients with neuroendocrine tumors of different types: clinical role. Ann Oncol. 2003;14:1135-1141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
60.  Zatelli MC, Torta M, Leon A, Ambrosio MR, Gion M, Tomassetti P, De Braud F, Delle Fave G, Dogliotti L, degli Uberti EC. Chromogranin A as a marker of neuroendocrine neoplasia: an Italian Multicenter Study. Endocr Relat Cancer. 2007;14:473-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 110]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
61.  Rodrigues M, Gabriel M, Heute D, Putzer D, Griesmacher A, Virgolini I. Concordance between results of somatostatin receptor scintigraphy with 111In-DOTA-DPhe 1-Tyr 3-octreotide and chromogranin A assay in patients with neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2008;35:1796-1802.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
62.  Belli SH, Oneto A, Aranda C, O’Connor JM, Domenichini E, Roca E, Méndez G, Bestani MC, Parma P, Giacomi N. Chromogranin A as a biochemical marker for the management of neuroendocrine tumors: a multicenter study developed in Argentina. Acta Gastroenterol Latinoam. 2009;39:184-189.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Marotta V, Nuzzo V, Ferrara T, Zuccoli A, Masone M, Nocerino L, Del Prete M, Marciello F, Ramundo V, Lombardi G. Limitations of Chromogranin A in clinical practice. Biomarkers. 2012;17:186-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]