Diagnosis and management of parathyroid carcinoma: a state-of-the-art review

in Endocrine-Related Cancer
Authors:
Pia Roser Department of Endocrinology ASO/EASO COM, King′s College Hospital NHS Foundation Trust, London, UK
Department of Endocrinology and Diabetes, University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Search for other papers by Pia Roser in
Current site
Google Scholar
PubMed
Close
,
Bianca M Leca University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK

Search for other papers by Bianca M Leca in
Current site
Google Scholar
PubMed
Close
,
Claudia Coelho Department of Diabetes and Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK

Search for other papers by Claudia Coelho in
Current site
Google Scholar
PubMed
Close
,
Klaus-Martin Schulte Department of Endocrine Surgery, King's College Hospital NHS Foundation Trust, London, UK
Academic Department of Surgery, Australian National University, Canberra, Australia

Search for other papers by Klaus-Martin Schulte in
Current site
Google Scholar
PubMed
Close
,
Jackie Gilbert Department of Endocrinology ASO/EASO COM, King′s College Hospital NHS Foundation Trust, London, UK

Search for other papers by Jackie Gilbert in
Current site
Google Scholar
PubMed
Close
,
Eftychia E Drakou Department of Clinical Oncology, Guy's Cancer Centre - Guy's and St Thomas' NHS Foundation Trust, London, UK

Search for other papers by Eftychia E Drakou in
Current site
Google Scholar
PubMed
Close
,
Christos Kosmas Department of Medicine, Division of Medical Oncology-Hematopoietic Cell Transplant Unit, Metaxa Memorial Cancer Hospital, Piraeus, Greece

Search for other papers by Christos Kosmas in
Current site
Google Scholar
PubMed
Close
,
Ling Ling Chuah Department of Endocrinology ASO/EASO COM, King′s College Hospital NHS Foundation Trust, London, UK

Search for other papers by Ling Ling Chuah in
Current site
Google Scholar
PubMed
Close
,
Husam Wassati Department of Radiology, King’s College Hospital NHS Foundation Trust, Princess Royal University Hospital, London, UK

Search for other papers by Husam Wassati in
Current site
Google Scholar
PubMed
Close
,
Alexander D Miras Section of Investigative Medicine, Division of Diabetes, Endocrinology & Metabolism, Imperial College London, London, UK

Search for other papers by Alexander D Miras in
Current site
Google Scholar
PubMed
Close
,
James Crane Department of Endocrinology ASO/EASO COM, King′s College Hospital NHS Foundation Trust, London, UK

Search for other papers by James Crane in
Current site
Google Scholar
PubMed
Close
,
Simon J B Aylwin Department of Endocrinology ASO/EASO COM, King′s College Hospital NHS Foundation Trust, London, UK

Search for other papers by Simon J B Aylwin in
Current site
Google Scholar
PubMed
Close
,
Ashley B Grossman Green Templeton College, University of Oxford, Oxford, UK
Centre for Endocrinology, William Harvey Institute, Barts and the London School of Medicine, London, UK
Neuroendocrine Tumour Unit, Royal Free Hospital, London, UK

Search for other papers by Ashley B Grossman in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0003-1176-6186
, and
Georgios K Dimitriadis Department of Endocrinology ASO/EASO COM, King′s College Hospital NHS Foundation Trust, London, UK
Obesity T2D and Immunometabolism Research Group, Faculty of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
Division of Biomedical Sciences - Reproductive Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, UK

Search for other papers by Georgios K Dimitriadis in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-6662-804X

Correspondence should be addressed to G Dimitriadis: georgios.dimitriadis@kcl.ac.uk

*(A B Grossman and G K Dimitriadis contributed equally to this work)

Open access

Sign up for journal news

Parathyroid carcinoma is one of the least common endocrine malignancies and accounts for approximately 1% of all patients with primary hyperparathyroidism. A systematic review of peer-reviewed literature published between January 2000 and March 2022 via Medline, Embase, Cochrane Central Register of Controlled Trials, EudraCT, ClinicalTrials.gov, CINAHL and SCOPUS was conducted. Manuscripts were eligible if they included data on adult non-pregnant populations with parathyroid carcinoma. No restrictions regarding interventions, comparators or duration of follow-up were imposed. Single case reports, reviews or meta-analyses were excluded. Outcomes of interest were molecular pathogenesis, clinical presentation, differential diagnosis, treatment, follow-up and overall survival. Study quality was evaluated using the Newcastle–Ottawa Scale for observational studies.

This review included 75 studies from 17 countries, reporting on more than 3000 patients with parathyroid carcinoma. CDC73 mutation has been recognised as playing a pivotal role in molecular pathogenesis. Parathyroid carcinoma typically presents with markedly increased calcium and parathyroid hormone levels. The most frequently described symptoms were bone and muscle pain or weakness. En bloc resection remains the gold standard for the surgical approach. The 5-year overall survival ranged from 60 to 93%, with resistant hypercalcaemia a significant cause of mortality. Emerging evidence indicating that targeted therapy, based on molecular biomarkers, presents a novel treatment option. The rarity of PC and need for personalised treatment warrant multidisciplinary management in a ‘centre of excellence’ with a track record in PC management.

Abstract

Parathyroid carcinoma is one of the least common endocrine malignancies and accounts for approximately 1% of all patients with primary hyperparathyroidism. A systematic review of peer-reviewed literature published between January 2000 and March 2022 via Medline, Embase, Cochrane Central Register of Controlled Trials, EudraCT, ClinicalTrials.gov, CINAHL and SCOPUS was conducted. Manuscripts were eligible if they included data on adult non-pregnant populations with parathyroid carcinoma. No restrictions regarding interventions, comparators or duration of follow-up were imposed. Single case reports, reviews or meta-analyses were excluded. Outcomes of interest were molecular pathogenesis, clinical presentation, differential diagnosis, treatment, follow-up and overall survival. Study quality was evaluated using the Newcastle–Ottawa Scale for observational studies.

This review included 75 studies from 17 countries, reporting on more than 3000 patients with parathyroid carcinoma. CDC73 mutation has been recognised as playing a pivotal role in molecular pathogenesis. Parathyroid carcinoma typically presents with markedly increased calcium and parathyroid hormone levels. The most frequently described symptoms were bone and muscle pain or weakness. En bloc resection remains the gold standard for the surgical approach. The 5-year overall survival ranged from 60 to 93%, with resistant hypercalcaemia a significant cause of mortality. Emerging evidence indicating that targeted therapy, based on molecular biomarkers, presents a novel treatment option. The rarity of PC and need for personalised treatment warrant multidisciplinary management in a ‘centre of excellence’ with a track record in PC management.

Introduction

Parathyroid carcinoma (PC) is a rare endocrine malignancy which accounts for less than 1% of all cases of primary hyperparathyroidism (PHPT) (Ruda et al. 2005). The incidence of PC is increasing, probably secondary to wider routine serum calcium screening (Lee et al. 2007). Hypercalcaemia is caused by increased and unregulated secretion of parathyroid hormone (PTH). Compared with patients with benign parathyroid adenomas (BPA), patients with PC are more symptomatic and present with more significantly raised calcium and PTH concentrations. PC is notable for the fact that it is challenging to diagnose, with confirmation typically only being possible post-operatively on histopathology. Certain histopathological features are suggestive of PC, but definitive diagnosis depends on the presence of invasion into surrounding tissues or distant metastasis (Lloyd et al. 2017). Surgery is the mainstay of initial treatment, as well as for local recurrence and distant metastases. There is a lack of data on the effectiveness and appropriate timing of radiotherapy (RT), while other treatment options such as chemotherapy or immunotherapy are limited (Wei & Harari 2012). The major morbidity of PC usually results from intractable hypercalcaemia induced by inoperable recurrent or metastatic lesions. Medical treatment is for symptomatic benefit and directed at managing such hypercalcaemia. Mutation of CDC73 has been recognised as playing an important role in molecular pathogenesis and has been associated with the predisposition to certain sporadic PCs, as well as hereditary syndromes such as hyperparathyroidism-jaw tumour syndrome (HPT-JT) (Gill 2014). Moreover, deeper understanding of the genetic landscape of PC can help determine the potential scope of new targeted therapy options.

Given the scarce data on PC, this study aims to systematically review the current evidence exploring the clinical manifestations, diagnosis and treatment of PC and provides recommendations to aid clinicians in the assessment and management of this rare condition.

Materials and methods

Protocol and registration

The protocol was developed according to the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols (PRISMA-P) and followed methods outlined in The Cochrane Handbook for Systematic Reviews of Interventions (Moher et al. 2015). This systematic review has been registered with PROSPERO (International Prospective Register of Systematic Reviews – University of York) with registration number CRD42020223157.

Eligibility criteria

Given the rarity of PC, the restrictions on the inclusion criteria were limited. Manuscripts in English, German and French, published between January 2000 and March 2022, were included for analysis. The search was conducted from 2000 onwards to ensure that the most recent relevant literature was included. Single case reports, expert opinion manuscripts, letters to the editor, commentaries, conference papers, animal studies, reviews and meta-analyses were excluded, while all other articles were included. Articles were eligible if they included data on non-pregnant, adult populations with PC. No restrictions regarding type of setting, stage of disease, exposures, interventions or duration of follow-up were imposed. Where available, comparison with benign parathyroid disease was acknowledged. Outcomes of special interest were related to molecular pathogenesis, clinical presentation, diagnosis, differential diagnosis, treatment, follow-up and overall PC survival.

Information sources and search methods

Systematic searches of the Medical Literature Analysis and Retrieval System Online (Medline), Excerpta Medica (Embase), Cochrane Central Register of Controlled Trials (CENTRAL), European Union Drug Regulating Authorities Clinical Trials (EudraCT), ClinicalTrials.gov, Cumulative Index of Nursing and Allied Health Literature (CINAHL) and SCOPUS databases between 2000 and 2022 were conducted. The search terms included parathyroid carcinoma or neoplasm, or cancer, or malign-, and the search strategy for MEDLINE was constructed as follows: (parathyroid(Title/Abstract)) AND ((carcinoma(Title/Abstract)) OR neoplasm(Title/Abstract)) OR cancer(Title/Abstract)) OR malign*(Title/Abstract))). The reference lists and other literature sources were browsed to ensure literature saturation. The final search was conducted in March 2022.

Study selection and data collection process

Study selection was performed independently and in parallel by two reviewers, against the set inclusion criteria and using the systematic reviews software Covidence (Veritas Health Innovation, Melbourne, Australia; http://www.covidence.org). Title and abstract were reviewed for relevance, followed by full-text assessment. Data on the existing evidence around incidence, epidemiology and risk factors, pathogenesis and histopathology, gene expression, clinical presentation, diagnosis, differential diagnosis, treatment options and outcomes, prognostic factors, follow-up and overall survival of PC were extracted from the selected articles independently by the two reviewers, using Microsoft Office Excel 2019. All disagreements encountered in the process of selecting articles and extracting data were resolved by consensus or arbitration by a third reviewer, as appropriate.

Study quality

Study quality assessment was performed independently by two reviewers using the Newcastle–Ottawa Scale (NOS) for observational studies. Studies were evaluated on eight items and categorised into three groups: selection (including whether the cohort is representative of the population), comparability (assessed on the grounds of study design and the analysis performed) and outcome (i.e. the assessment of outcome, follow-up rate and adequacy follow-up period). For each question, there are several possible answers, using a star system to allow a semi-quantitative assessment of study quality. A study can be awarded a maximum of one star for each numbered item within the selection and outcome categories. A maximum of two stars can be given for Comparability, with between zero and nine stars attainable (Stang 2010). The scores of the articles using the NOS were translated to the Agency for Healthcare Research and Quality (AHRQ) standards (good, fair and poor), using the conversion thresholds developed by the AHRQ.

Data synthesis

Heterogeneity was visually inspected but, due to high variability and insufficient data, results were not pooled into a meta-analysis. A narrative synthesis was therefore conducted instead.

Results

Study selection

The search retrieved a total of 3097 studies. After removing duplicates and reviewing abstracts, 326 full-text articles were examined in detail for eligibility. The findings from 75 articles have been included in our systematic review. The PRISMA flow diagram is shown in Fig. 1 (Moher et al. 2015).

Figure 1
Figure 1

Study selection flowchart. The search retrieved a total of 3097 articles. After removing duplicates and reviewing abstracts, 326 full-text articles were analysed in detail for eligibility. Around 77 studies met the eligibility criteria and 75 studies were included in the review. A full colour version of this figure is available at https://doi.org/10.1530/ERC-22-0287.

Citation: Endocrine-Related Cancer 30, 4; 10.1530/ERC-22-0287

Study characteristics

Totally 75 studies were systematically reviewed. Of these, 22 studies were laboratory studies. The remaining studies were observational studies (case series, case–control and cohort studies, n = 52) and one single-arm prospective study. In total, 38 studies were single-centre, 4 were multicentric and 11 used national databases. It is not possible to report the exact total number of participants with PC described in this paper, as there may be a significant overlap in patient populations given the inclusion of national databases. We report on more than 3000 patients with avoidance of possible overlapping cases through inclusion of only one database at a time when the same database was used in different studies. The sample size ranged from 2 to 1022 patients. Patients were included from 17 different countries. In 21 studies, PC findings were compared to benign disease. The studies informed on different and multiple aspects of PC management – 28 stated clinical manifestations, 42 follow-up and 29 survivorship from PC. Furthermore, two studies proposed and validated prognostic classification systems. Table 1 and 2 in the main manuscript and Supplementary Table 1 (see section on supplementary materials given at the end of this article) summarise each of the selected studies looking at the period of review, setting/country, sample size and key findings from each study.

Table 1

Study characteristics of observational studies included in this systematic review (between 2000 and 2022) investigating diagnosis and management of parathyroid carcinoma.

Ref Review period Setting PC sample size Total sample size Quality M/F (n) Age in years: mean/median PTH (pg/mL): mean/median Serum total Ca (mg/dL): mean/median Ca(2+) (mmol/L): mean/median Serum ALP (IU/L): mean/median Serum Phosphate (mg/dl): mean/median Tumor size (mm): mean/median Differences PC vs BPA/APN/PH
Observational studies: single centre
Apaydin et al. (2021) 2012–2020 Istanbul, Turkey 7 540 9 2/5 NR/47 (IQR, 40–51) 1115/NR (IQR, 287–1470) NR/12.7 (IQR, 11.3–13.9) NR NR/367 (IQR, 214–1607) NR NR/29 (IQR, 28–40) NR
Bae et al. (2012) 2000–2011 Seoul, South Korea 11 194 9 7/4 54±14/NR NR/1000 (IQR, 467 – 1714) 13.1±3.4/NR NR NR/541 (IQR, 232–1157) 3.2±2/NR 38±16/NR Higher Ca, +ALP, PTH, +P levels, larger tumor size
Basceken et al. (2015) 2000–2012 Ankara, Turkey 11 522 8 7/4 49±14/NR NR NR NR NR NR NR NR
Busaidy et al. (2014) 1980–2003 Houston, USA 27 27 9 16/11 47±15/NR (R, 16–75) NR 13.4±1.9/NR (R, 10.4–17.7) – n=18 NR NR NR 30/NR NR
Chen et al. (2003) 1987–2001 Kobe, Japan 20 131 7 9/11 50±3/NR 397±138/NR 12.7±0.4/NR NR 845±243/NR 2.3±0.1/NR NR Higher Ca, ALP and PTH levels, lower age, lower BMD in the radius
Chen et al. (2018) 2013–2016 Nanjing, China 4 4 7 2/2 53±3/NR 1557±851/NR (R, 434–2480) 13.3±2.8/NR NR 267±203/NR (R, 81–559) NR NR/35±10 (R, 1.9–4 cm) NR
Christakis et al. (2016b) 1980–2001; 2002–2015 Houston, USA 26 ; 31 26 ; 31 9 18/8 ; 12/19 45±9 /NR (R, 28–54); 53±13/NR (R, 13–79) NR 13.9±2.1 (R, 10.2–17.7)/NR (n=14) 13.2±2.3 (R, 9.7–20.5)/NR (n=22) NR 169±75 (R 71–255) (n=9); 183±226 (R 23–343) 2.7±0.3 (R 2.2–3.2) (n=10); 3.1±0.8 (R 2.1–3.9) (n=5) 23±8/NR; 28±10/NR NR
Christakis et al. (2016a) 2000–2014 Houston, USA 31 54 9 18/13 54±11/NR (R, 28–77) NR/444.5 (n=34) NR /12.3 (n=34) NR/1.7 (n=18) NR NR 24/NR Higher PTH, more male pat. in PC
Christakis et al. (2017a) 1980–2015 Houston, USA 8 8 8 6/2 NR/53 NR NR NR NR NR NR/28 (R, 11–40) NR
Christakis et al. (2017b) 2000–2015 Houston, USA 20 20 7 11/9 NR NR NR NR NR NR NR NR
Erovic et al. (2012) 1976–2005 Toronto, Canada 16 16 7 6/10 54/54 (R, 31–72) 1238/1103 (R, 145–3112) 19.9/15.8 (R, 9.7–48.9) NR NR NR 33/28 (R, 15–75) NR
Harari et al. (2011) 1966–2009 San Francisco, USA 37 37 8 23/14 53/NR (R, 23–75) NR 14.1/NR (R, 9.8–24) NR NR NR 31/NR (R, 18–100) NR
Hu et al. (2019) 1997–2018 Beijing, China 53 53 9 35/18 46±13/NR (R, 24–71) 1288±805/NR 13.7±2.1/NR NR NR NR 33±13/NR NR
Iacobone et al. (2004) 1980–2000 Padua, Italy 19 404 8 12/7 60±14/65 (R, 30–78) 506±417/NR 13.4±1.9/NR NR 188±108 NR 31±9/NR Lower female:male ratio, larger tumor size, higher Ca and PTH levels in PC
Iihara et al. (2007) 1981–2005 Tokyo, Japan 38 38 9 17/21 NR/51 (R, 13–74) Times the UNL 1.8–33 13.8/NR (R 11.3 – 20) NR NR NR NR NR
Karakas et al. (2012) 1987– 2008 Marburg, Germany 19 1363 8 12/7 49±12/NR 1250±769/NR 15.2±1.2/NR NR 408 0.7 NR Younger age, higher Ca and PTH levels in pat. with PC vs.pHPT
Kirkby-Bott et al. (2005) 1991–2002 London, UK 7 7 8 5/2 NR/44 (25–70) NR NR/13.9a NR NR NR NR NR
Kleinpeter et al. (2005) 1975–2004 Winston–Salem, USA 23 23 8 9/14 54/NR NR/290 (R, 96–1542) 12.9/NR (R, 8.7–21.9) NR NR NR NR NR
Kowalski et al. (2022) 1983–2019 Katowice, Poland 29 921 9 12/17 55/NR (R, 27–77) 313.4±381.1/NR NR/NR 2.03±0.39/NR NR 1.08±0.31 24.8/NR (R, 15 – 65) Higher PTH, Ca, urea and creatinin levels in pat. with PC vs. BPA
Lee et al. (2010) 1992–2007 Seoul, Korea 7 171 9 2/5 43/NR (R, 23–60) 620.1 pg/ml (R, 50.6–1417) 11.4/NR (R, 10.3–12.3) NR NR NR NR NR
Liu et al. (2021) 2004–2020 Beijing, China 21 21 7 9/12 NR/45 (R, 24–71) NR/345 (R, 68.8–3304) NR/12.38 (R, 9.6–18.8) NR NR NR NR/18 (R, 8–38) NR
Medas et al. (2016) 2001– 2014 Cagliari, Italy 8 319 8 1/7 62±6/NR (55–73) 547±655/NR (122–1693) 11.9±2.1/NR (9.2–15.6) NR NR NR NR NR
Munson et al. (2003) 1928–1999 Minnesota, USA 61 61 8 26/35 53/52 NR 14.4/13.8 NR NR NR NR NR
O’Neill et al. (2011) 1999–2010 Sidney, Australia 7 1292 9 6/1 57/NR 609/NR 13.1/NR NR NR NR 28/NR More males and higher tumor weight in PC (in univariate analysis p<0.05), higher Ca, PTH and Tumor size, p<0.05 in multivariate analysis in pat. with PC vs. PHPT
De Pasquale et al. (2021) 2011–2021 Milan,Itlay 8 462 8 5/3 58/NR (R, 28–78) NR/736 (205–4349) NR/13.7 (R 12,5–19,3) NR NR NR/2.1±0.425 (R, 0.6–3.3) NR/23 (R 13–36) More frequently male and higher preoperative Ca and PTH + lower phosphate levels, larger and heavier parathyroids excised, lower postoperative Ca, and a higher rate of postoperative hypoparathyroidism in pat. with PC vs. PHPT
Quinn et al. (2015) 2001–2014 Yale, USA 18 3643 8 8/10 59/NR (R, 51–67) 489/NR (R, 240–739) 13.0/NR (R, 12.2–13.8) NR NR NR 35/NR (R, 27–42) higher Ca, PTH levels, increased tumor size in pat. with PC vs. APTA
Robert et al. (2005) 1976–2001 Geneve, Switzerland 9 311 8 2/7 55±10 (R, 35–67)/57 (IQR 11) Upper R of Normal Value 10±7 (4–20)/6.8 (IQR 9.3) 13.6±4.4 (R, 10.4–24)/13.2 ( IQR, 3.2) NR NR NR NR Higher Ca, PTH levels, higher tumor weight in pat. with PC vs. pHPT
Sali et al. (2021) 2004–2018 Mumbai, India 20 20 7 10/10 NR/49 (R, 21–73; IQR, 42–57) 864.5 (12/20)/NR 13.51 (13/20)/NR NR NR NR 24±8.8 (14/20)/NR NR
Schulte et al. (2014) 1996– 2013 London, UK 19 19 8 R0: 3/9 R1: 2/5 R0: 53.1±11.3/57.5 (R, 33–67) R1: 65.7±9.5/59 (R, 54–81) Times the UNL R0: 9.5±8.0/4.7 (R, 1.5–22.3) R1: 7.1± 5.0/5.7 (R, 1.3–13.7) Times the UNL R0: 12,42±2.4/11.2 (11.2–18.8); R1: 12,42± 1.2/11.6 (10.8–14.4) NR NR NR R0: 38.7±17.4/35 (R, 15–65) R1: 32.7±14.7/30 (R, 20–56) NR
Schulte et al. (2010) 2005–2010 London, UK 11 11 8 5/6 55/NR (R, 34–70) 556/NR (R, 116–1295) 13.2a (R, 10.4–18.8)/NR NR NR NR NR NR
Schulte et al. (2021) 1980–2018 Chicago, USA 6 27 8 NR NR 957/NR (R, 79–2250) 11.8/NR (R, 11–14.2) NR NR NR 28/NR (R, 13–37) No significant difference of gland size/weight, serum PTH/Ca levels, and locoregional recurrence. PC was more likely to display coarse chromatin with nucleoli, infiltrative invasion and metastasis. Infiltrative invasion was more common in cases with progression and metastasis.
Selvan et al. (2013) 1988–2008 Vellore, India 10 245 7 7/3 52/NR (20–87) 1111/NR 15.1/NR NR NR NR 23±2/NR NR
Shah et al. (2021) 2010–2018 Western India 19 112 9 10/9 46±15/NR NR/1098 (IQR, 565–1729) 13.8±2.3/NR NR NR/527 (IQR, 135–1075) 2.3±0.9/NR (R, 2.5–4.5) 34.6±11.5/NR Longer tumor diameter, higher short/long axis, more irregular shape, infiltration, heterogeneity, calcification, older age, higher Ca levels in pat. with PC
Sidhu et al. (2011) 2004–2009 London, UK 8 69 8 NR NR/56 (IQR, 37–64; R, 20 – 70) UNL NR/5.6 (IQR, 2.8–14.1; R, 1.7–20.9) NR/12.0 (IQR, 10.8 – 14.4; R, 10.8 – 18.8) NR NR NR NR/38.5 (IQR:20 – 62.5; R, 20.0 – 75.0) Higher serum Ca, PTH, tumor size in pat. with PC
Wächter et al. (2019) 1985–2018 Marburg, Germany 18 1705 9 11 /7 NR/46 (R, 17–66) NR/1110 (R, 489–3100) NR/15.6 (R, 13.2 – 17.6) NR NR NR NR NR
Wei et al. (2022) 2010–2019 Beijing, China 31 31 7 20 /11 46 ±14/NR EEBR with vs. without distant metastasis: 921.3 vs 344.2 EEBR with vs. without distant metastasis: 14.47 vs 11.74 NR NR NR NR NR
Xue et al. (2016) 2000–2015 Shangai, China 40 322 9 17/23 49±2/NR NR/1622 (R, 200–3251) 14.1±3.9/NR NR NR/234 (R, 64–2786) NR 32.5±10/NR Higher Ca, PTH and parathyroid volume in pat. with PC
Observational studies: multicentric
Young et al. (2016) 1999–2012 California Cancer Registry, USA 136 136 8 71/65 57±13/NR NR NR NR NR NR NR/25 (IQR, 17 – 35) NR
Schulte et al. (2012) NR 8 centres in USA, Australia, UK, Poland, Germany, Japan, Italy 82 82 8 40/42 52±15/51 (R, 25–84) UNL 14.2±13.8/10.7 (R 1.2–86) 13.6±2.0/13.6 (R, 9.2 –18.8) NR NR NR 35±18/30 (R, 15–120) NR
Lenschow et al. (2022) 1986–2008 29 centres in Germany, Austria, Switzerland 83 83 7 45/38 NR/54 (R, 22 – 94) NR/566 (R, 31.8 – 8900) NR/13.4 (R, 9.2 – 24) NR NR NR NR NR
Villar del Moral et al. (2014) 1980–2013 35 centres in Spain 62 6863 7 38/24 NR/59 (IQR, 51–70) NR/507 (IQR, 225–934) NR/13a (IQR, 11.6–14.6) NR NR NR NR/28 (IQR, 20–38) NR
Observational studies: national database analysis
Asare et al. (2015) 1985–2006 NCD, USA 733 733 8 327/406 56±15/57 (R, 15–89) NR NR NR NR NR 29.6±18.4/25 (R, 10–150) NR
Hsu et al. (2014) 1988–2010 SEER database, USA 405 405 8 212/193 NR/56 (IQR, 46 – 66) NR NR NR NR NR NR/28 (IQR, 19–38) NR
Kong et al. (2021) 2002–2017 NHIS, Korea 255 255 9 100/155 53±16/NR NR NR NR NR NR NR NR
Leonard-Murali et al. (2021) 2004–2015 NCD, USA 555 555 8 286/269 NR NR NR NR NR NR NR NR
Limberg et al. (2021) 2004–2016 NCD, USA 885 885 8 No EBRT: 395/364 EBRT: 71/55 No EBRT: 58±14/NR EBRT: 56±12/NR NR NR NR NR NR No EBRT/EBRT <30mm – 292/46 >=30mm – 199/47 NR
Observational studies: national database analysis
Lo et al. (2018) 1973–2014 SEER database, USA 520 520 8 267/253 NR/57 (R, 20 – 87) NR NR NR NR NR 0–29 – 176 ≥30 – 133 NA – 211 NR
Qian et al. (2022) 2001–2018 SEER database, USA 604 604 9 311/293 NR/59 (IQR, 48–68) NR NR NR NR NR <=20 – 125 >20,<=40 – 189 >40, <=60 – 43 >60 – 20 NA – 227 NR
Ryhänen et al. (2017) 2000– 2011 Finland 32 132 9 14/18 NR/61 (R, 17–83) NR/989 (IQR, 461 – 1518) NR NR/1.76 (IQR, 1.61–1.97) NR/119 (IQR, 74–166) NR/0.8 (IQR, 0.63–1.04) NR/29.5 (IQR, 20 – 30) Higher caconcentrations, PTH levels, larger tumor size, higher alk. phosphatase, higher creatinine in pat. with PC
Sadler et al. (2014) 1998–2011 NCD, USA 1022 1022 8 505/517 57 ±14/57 NR NR NR NR NR NR NR
Shah et al. (2021) 2010–2018 Western India 19 112 9 10/9 46±15/NR NR/1098 (IQR, 565–1729) 13.8±2.3/NR NR NR/527 (IQR, 135–1075) 2.3±0.9/NR 34.6±11.5/NR Longer tumor diameter, higher short/long axis, older age, higher Ca concentrations, heterogeneity, infiltration, calcification, irregular shape in pat. with PC
Zhou et al. (2021) 1973–2015 SEER database, USA 593 593 8 299/295 NR NR NR NR NR NR NR NR
Interventional study: open-label, single-arm, dose titration
Takeuchi et al. (2017) NR Japan 7 7 7 4/3 66±8/NR (R, 51–74) 434±380/NR (R, 181–1230) 12.1±1.3/NR (R, 10.4–14.6)a NR NR 2.2±0.6 mg/dL NR NR

aAlbumin-corrected serum Ca

ALP, alkaline phosphatase; APN, atypical parathyroid neoplasm; BMD, bone mineral density; BPA, benign parathyroid adenoma; CND, central neck dissection; CSMR, cancer-specific mortality rate; DFS, disease-free survival; DSS, disease-specific survival; EBRT, external beam radiotherapy; EEBR, extended en bloc resection; HTx, hemithyroidectomy; ILND, ipsilateral lymph node dissection; IQR, interquartile range; LN, lymph node; LR, local resection; MIPC, minimally invasive parathyroid carcinoma; MS, median survival; NA, not assessed; NCD, National Cancer Database; NHIS, National Health Insurance Services; NR, not reported; OMR, overall mortality rate; OS, overall survival; OS, overall survival; PC, parathyroid carcinoma; PC, parathyroid carcinoma; PH, parathyroid hyperplasia; PTH, parathyroid hormone; PTx, parathyroidectomy; R, range; SEER, surveillance, epidemiology, and end results; sTx, subtotal thyroidectomy; Tx, total thyroidectomy; UNL, upper normal limit; WIPC, widely invasive parathyroid carcinoma; Y, years.

Table 2

Study characteristics of observational studies included in this systematic review (between 2000 and 2022) investigating diagnosis and management of parathyroid carcinoma.

Ref Treatment Radiotherapy (n) FU time in months: mean/median Recurr. (n) Initial metast. (n) Survivorship Prognostic Factors
Observational studies: single centre
Apaydin et al. (2021) PTx + (within 3 months after) sTx or HTx-3 PTx + HTx-4 4 NR/60 (IQR, 29–75) NR 0 NR NR
Bae et al. (2012) NR NR 56/NR NR NR NR NR
Basceken et al. (2015) PTx-5

PTx + HTx-1

PTx +HTx + ICND-2

PTx + TT-1

PTx + TT + CND-2
NR 100±42/NR 0 1 91% OS (5 y) NR
Busaidy et al. (2004) PTx-18

En-bloc resection (PTx + HTx, excision of the strap musculature, and adjacent soft tissues as required)-9
6 95/NR 11 1 85% OS (5 y)

77% OS (10 y)
No differences in survival by sex, age, tumor size or calevels.
Chen et al. (2003) NR NR NR NR NR NR NR
Chen et al. (2018) PTx-1

PTx + ipsilateral HTx-3
NR NR 1 NR NR NR
Christakis et al. (2016b) PTx-10; 9

PTx and ipsilateral HTx-3; 10

PTx and ipsilateral HTx with CND-4; 9

PTx with CND-2; 2
7; 4 NR/144 (R, 0–326); NR/35 (R, 1–145) 19 NR 82%; 72% OS (5 y) 62%; 66% DFS (5 y) NR
Christakis et al. (2016a) PTx-6

PTx and HTx-5

PTx, HTx and CND-8
NR NR/13 (n=24) 10 NR 83%; 76%; 67% OS (5, 10, 15 y)

60% DFS (5 y)
NR
Christakis et al. (2017a) PTx-2

PTx and HTx-2

PTx, HTx and CND-4
8 NR/150 (R, 24–240) 4 NR NR NR
Christakis et al. (2017b) NR NR 58±66/NR NR NR NR NR
Erovic et al. (2012) PTx-7

PTx + sTx-6

Tx-1

Tx + neck dissection-1

PTx + thymectomy + neck dissection-1
11 88/79 (R, 2–153) 7 NR 69%; 43% DFS (5, 10 y)

100%; 80% DSS (5, 10 y)
NR
Harari et al. (2011) NR NR 58/NR 18 NR 78% OS (5 y)

67% OS (10 y)
Associated with increased mortality: lymph node or distant metastases, number of recurrences, higher Ca at recurr., and a high number of calowering agents.
Hu et al. (2019) LR-35

En-bloc ressection (LR and ipsilateral HTx)-18 + CND (11/18)
2 NR/80 (R, 7–282) 32 1 90%; 78% OS (5, 10 y)-

36%; 32% DFS (5, 10 y)
Related to mortality: age > 50 years. Neg parafibromin staining was related to recurrence and metastases.
Iacobone et al. (2004) En-bloc resection (PTx, ipsilateral HTx and thymectomy)-19 1 NR/29 (R, 20–153) 13 NR mDFI-15 months (R, 2 – 74 months)

MS-29 months (R, 20 – 146 months)
Overall survival was significantly correlated with the disease-free interval.
Iihara et al. (2007) LR-16

En-bloc resection (LR + ipsilateral HTx)-22 + LND-4/22
NR NR/119 (R, 14–396) 15 2 90%; 86% DSS (5, 10 y) Locoregional tumor extension affected cause-specific survival (P = 0.05), Ki-67 ≥5% decreased disease-free survival (P = 0.005).
Karakas et al. (2012) PTx-10

En-bloc resection (resection of all tumor-bearing tissue + HTx + CND)-9
NR NR/60 (R, 27–243) NR 1 93%; 43% OS (5, 10 y) NR
Kirkby-Bott et al. (2005) PTx-5

En-bloc resection of parathyroid tumor with HTx, hemi-thymectomy and CND-2
5 NR 3 NR NR NR
Kleinpeter et al. (2005) PTx-11

En-bloc resection (PTx + concurrent ipsilateral thyroid lobectomy+excision of adherent strap musculature, the parathracheal fibrolymphatic tissue + thymus if involved)-12
2 NR/134 (R, 1–276) 5 NR 86%; 69% OS (5, 10 y)

90%; 68% En-bloc OS (5, 10 y)

83%; 71% LR OS (5, 10 y)
NR
Kowalski et al. (2022) En-bloc resection of the tumor along with the surrounding tissues-9

PTx with the removal of the ipsilateral HTx-8

LR-12
3 NR 9 6 69% OS (10 y) NR
Lee et al. (2010) PTx + ipsilateral HTx-7

PTx + Tx-1
0 103/NR (R, 36–254) 1 NR NR NR
Liu et al. (2021) PTx-15

En-bloc resection-6
NR NR 29° NR mDFS-36 (2-205) months NR
Medas et al. (2016) PTx-5

PTx + HTx-1

PTx + Tx-1

PTx + prophylactic CLND-1
0 NR 2 NR NR NR
Munson et al. (2003) NR 4 NR/76 (R, 8.4–358) 25 NR 77% OS (5 y)

82% DSS (5 y)
Surgical margin status is a predictor of locoregional disease progression. Surgical institution was a significant independent predictor of DSS.
O’Neill et al. (2011) NR NR NR/44 (R, 6–78) NR NR NR NR
De Pasquale et al. (2021) En-bloc resection (PTx + ipsilateral HTx + superior PTx )-3

En-bloc resection (PTx + Tx + superior PTx + CLND)-2

LR-3
NR 38.4/NR (R, 13–109) 0 0 NR NR
Quinn et al. (2015) Radical PTx-9 NR NR 6 2 NR NR
Robert et al. (2005) NR NR NR/96 (R, 3–240) 0 NR NR NR
Sali et al. (2021) En-bloc resection of the involved parathyroid gland-16 8/16 (50%) 24/21.5 (R, 6–58) (16/20) 2 NR NR NR
Schulte et al. (2014) LR-6 (R1-5, R0-1)

Oncologic resection (En-bloc resection of the parathyroid lesion with surrounding fat tissue, ipsilateral thyroid lobe, and level VI lymph nodes)-13 ( R1-2, R0-11)
3 NR 5/7 in R1 NR NR R0 margins and primary oncological resections were associated with higher DFS (P = 0.005 and P = 0.03).
Schulte et al. (2010) NR NR NR/15 (R, 1–27) 1 1 NR NR
Schulte et al. (2021) NR NR 95/NR (R, 8–293) 1 NR NR NR
Selvan et al. (2013) Inadequate resection= HTx and excision of the tumor-3

En-bloc resection = ipsilateral HTx, central node clearance, and soft tissue resection, which included strap muscles and prevertebral fascia-7
6 NR/43 3 NR NR NR
Shah et al. (2021) Radical surgical excision incl. PTx with ipsilateral HTx with/without lymph node clearance-9

Isolated PTx-10
NR 29±14/NR NR NR NR NR
Sidhu et al. (2011) NR NR NR NR NR NR NR
Wächter et al. (2019) PTx-9

En bloc resection (PTx + ipsilateral HTx + ipsilateral CND)-9
NR NR/108 (R, 41–316) 10 3 100%; 56% PTx-OS (5, 10 y)

78%; 33% PTx + HTx + ipsilateral CND-OS (5, 10 y)
The recurrence-free survival after PTX+HTX was significantly longer than after PTX (143 vs. 18 months, P = 0.01), while the overall survival of both groups after a median follow-up of 107.5 months did not significantly differ.
Wei et al. (2022) EEBR-25

LR-6
NR NR/27 (R, 10–101) 31 NR 60% vs. 17% EEBR vs Local Resection in patients with recurrent or persistent PC OS (5 y) UAT invasion and preoperative serum Ca were independent factor associated with an increased mortality rate. For patients with Upper aerodigestive tract (UAT) invasion, there was an associated with a 7.6 times increase in the risk of mortality.
Xue et al. (2016) LR-14 NR NR/49 (R, 2–177) 12 NR 79%; 61% OS (5, 10 y) Ca, PTH-Levels, local excision and recurrence were associated with mortality.
Observational studies: multicentric
Young et al. (2016) PTx-60

PTx + Tx-58

PTx with delayed thyroid

resection-18
NR NR/64 (IQR, 30–105) NR NR 87% OS (5 y)

72% OS (10 y)
No significant difference found in OS between en-bloc resection and PTx alone. Factors that were independently associated with decreased survival included age and distant metastases.
Schulte et al. (2012) PTx-32

En-bloc resection (PTx + circumferential soft tissue +-ipsilateral HTx, CND or further LR)-108
NR 76±74/50 (R, 2–347) 25 NR Only reported in subgroups Mortality was exclusive to the high-risk group, which also predicted a significant risk of recurrence (risk ratio 9.6; 95% confidence interval 2.4–38.4; P < 0.0001), with significantly lower 5-year disease-free survival (χ2 = 8.7; P < 0.005 for n = 45).
Lenschow et al. (2022) PTx-19

PTx and hTx-20

En-bloc resection (PTx+hTx+CND)-13

En-bloc resection + lLND 8 (9.6%)

PTx + Tx-8

PTx + Tx + CND-2

PTx + Tx + CND/lLND-4

Unknown-8

Two-stage completion operation-25
8 NR/50 (R, 0–460) (n=74) 32 19 90% OS (50 months)

98% DSS (50 months)
Recurrence war reduced when extended initial surgery had been performed. Lower T-status, N0 stage at initial diagnosis, Ki-67 <10% and postoperative biochemical remission were beneficial prognostic parameters for recurrence-free survival.
Villar del Moral et al. (2014) PTx-18

En-bloc resection-44
2 NR/52 (IQR, 16–97) 14 NR 92%; 69% DSS (5,10 y) As to disease-specific survival, poor outcomes were associated with intraoperative tumor rupture and distant recurrence.
Observational studies: national database analysis
Asare et al. (2015) LR, single/partial removal of tumor, local tumor destruction, debulking-444

Complete excision of the entire parathyroid mass-132

RS (Removal of the parathyroid tumor with concomitant resection (partial or total removal) of adjacent organs)-72

No surgery-28
51 NR/65 (R, <1–312) NR NR 82%; 66% OS (5,10 y) Patient age, tumor size, and sex have modest effects on survival in patients with parathyroid carcinoma.
Hsu et al. (2014) PTx-329

En-bloc resection-42

Debulking and local destruction procedures-8

None-12
37 NR/68 (IQR, 29–106) NR NR 3%; 65% OS (5,10 y)

94%; 90% DSS (5,10 y)
Tumor size ≥3 cm and distant metastasis were significant prognostic factors of DSS in PC. Metastatic lymph nodes did not independently predict worse DSS.
Kong et al. 2021 PTx + Tx-82

PTx-173
NR NR/68 (IQR, 42–113) 14 NR 87%; 73% OS (5,10 y) Age over 50 years, Tx at the initial surgery, and reoperation had increased risk of mortality.
Leonard-Murali et al. (2021) LR-522

RS-33 ( en bloc resection of the tumor and ipsilateral Tx, CND-/+ LND)
482 NR/60.4 NR 12 NR There was no difference in OS between LR and RS for localized PC. Age >75 years, unknown ethnicity, government insurance and Charlson/Deyo score of 1 and of ≥2 were associated with worse survival.
Limberg et al. (2021) No EBRT/EBRT

LR-429/65

Total excision-264/44

EEBR-39/10

Debulking or surgery, not otherwise specified-27/7
126 NR/60.8 NR No EBRT/EBRT-8/3

>=pN1 No EBRT/EBRT-10/10
85%; 67% OS (5,10 y) EBRT is not associated with any survival benefit in the treatment of PC.
Observational studies: national database analysis
Lo et al. (2018)

PTx-408

En-bloc resection-48

Refused surgery-47
45 NR/54 (n=42) NR 12 89% DSS

11% CSMR

median DSS-75
Patients with

metastatic disease and tumors > 3 cm have worse cancer-specific survival.
Qian et al. (2022) PTx-488

EEBR-51

Other-38

No surgery-27
64 NR/76 (IQR: 33–135) NR NR NR Age at diagnosis > 70 years, tumor size > 35 mm, and distant metastasis were independent risk factors for PC-specific mortality. Compared with distant metastasis, localized and regional lesions showed an improved CSS rate.
Ryhänen et al. (2018) En-bloc/radical resection-15 (PTx + ipsilateral Tx + adjacent structures or lymph nodes)

LR-17
7 NR/80 (R, 24–167) 6 NR 91% OS (5 y)

72% OS (10 y)
NR
Sadler et al. (2014) LR-567

Total removal-350

EEBR-53

Debulking-12
127 NR/77 NR NR 81% OS (5 y) (n=528) Positive lymph nodes and older age were associated with lower OS.
Shah et al. (2021) Radical surgical excision incl. PTx with ipsilateral HTx with/without lymph node clearance-9

PTx-10
NR 29±14/NR NR NR NR NR
Zhou et al. (2021) PTx-384

En-bloc resection-38

Debulking-5

No definitive treatment-40
57 NR NR NR MS-83 months (R 34–146.75)

10% CSMR,

35% OMR
The extent of resection was related to both CSS and OS, whereas race and extent of disease had a significant positive correlation with OS. Patients who underwent parathyroidectomy had remarkably better CSS and OS than patients who did not undergo definitive treatment. Metastatic disease was a significant predictor of OS.
Interventional study: open-label, single-arm, dose titration
Takeuchi et al. (2017) PTx-5 NR NR NR NR NR NR

° >=1 recurrence in each patient

ALP, alkaline phosphatase; APN, atypical parathyroid neoplasm; BMD, bone mineral density; BPA, benign parathyroid adenoma; CND, central neck dissection; CSMR, cancer-specific mortality rate; DFS, disease-free survival; DSS, disease-specific survival; EBRT, external beam radiotherapy; EEBR, extended en bloc resection; HTx, hemithyroidectomy; ILND, ipsilateral lymph node dissection; IQR, interquartile range; LN, lymph node; LR, local resection; MIPC, minimally invasive parathyroid carcinoma; MS, median survival; NA, not assessed; NCD, National Cancer Database; NHIS, National Health Insurance Services; NR, not reported; OMR, overall mortality rate; OS, overall survival; OS, overall survival; PC, parathyroid carcinoma; PC, parathyroid carcinoma; PH, parathyroid hyperplasia; PTH, parathyroid hormone; PTx, parathyroidectomy; R, range; SEER, surveillance, epidemiology, and end results; sTx, subtotal thyroidectomy; Tx, total thyroidectomy; UNL, upper normal limit; WIPC, widely invasive parathyroid carcinoma; Y, years.

Incidence

PC is rare, but its incidence appears to be increasing. Ryhänen et al. (2017) and Lo et al. (2018) assessed PC incidence. The latter evaluated the National Cancer Institute’s Surveillance, Epidemiology, and End Results Programme (SEER) database which encompasses approximately 28% of the U.S population. The annual overall incidence of PC increased from 2 to 11 cases per 10 million between 1974 and 2001 and has remained stable around 10–13 cases per 10 million per annum since 2001. Lo et al. (2018) showed in a subgroup analysis of significant prognostic factors that there was a trend towards increasing incidence of tumours between 0 and 3 cm and a trend towards increasing incidence of local vs distant disease at the time of initial operation. Likewise, in Finland, the annual incidence of PC has risen over the last decade, with 1.4 cases from 1955 to 2000 to 7.1 cases per 10 million from 2000 to 2013 (Ryhänen et al. 2017). A potential explanation is higher detection rates attributable to improved diagnostic methods and greater life-expectancy (Ryhänen et al. 2017).

PC is rare, but its incidence has increased over the last decades to 10–13 cases per 10 million per year, which could be explained by higher detection rates due to improved diagnostic methods and greater life-expectancy.

Diagnostic evaluation

Clinical presentation and complications at diagnosis

PC presents equally in males and females (Sadler et al. 2014, Ryhänen et al. 2017, Lo et al. 2018), unlike BPA, which is more common in females (Iacobone et al. 2004). The median age of presentation was 44–65 years, with the largest study of 1022 patients having a mean age of 57 years (Sadler et al. 2014). No significant correlations have been shown with ethnicity, geographical area or socio-economical level in the current literature. Patients with PC most commonly present with signs and symptoms such as bone and muscle pain, neuropsychiatric symptoms, kidney stones, hypercalcaemic crisis, osteopenia/osteoporosis, pathological fractures and a palpable neck mass. Kowalski et al. (2022) suggest the active consideration of PC when the patient presents both renal and bone symptoms simultaneously, because dual presentation is quite rare in BPA. It should be stated that the prevalence of bone and kidney involvement in BPA depends on the definition of ‘bone and kidney involvement’, and that hypercalciuria or the detection of microcalcification by kidney ultrasound as well as the occurrence of osteopenia, might also occur in patients with BPA.

The clinical presentation of patients from 27 studies, including 932 patients, is summarised in Supplementary Table 3. Bone manifestations were described most frequently with osteoporosis/osteopenia, fractures and bone plus muscle pain/weakness, being present in 45.8% (n = 427). Renal manifestations including kidney/urinary stones and renal failure were present in 37.2% (n = 347), followed by fatigue (13.6%, n = 127), a palpable neck mass (11.9%, n = 111) and neuropsychiatric symptoms (11.2%, n = 104). Of note, only 3% (n = 28) of patients were asymptomatic. We found that studies often did not have robust clinical presentation data and many symptoms were not commented upon.

Table 3

Study characteristics of laboratory studies included in this systematic review (between 2000 and 2022).

Reference Period of Review Setting PC Sample size Total Sample size Quality Key findings
Laboratory Studies
Agarwal et al. (2021) 2018−2020 New Delhi, India 7 126 8 Normal autopsy-derived parathyroid tissues were Her2/Neu 3 +, but incomplete membranous staining predominated in 85%. Their immune-scores were significantly more than the PC cases (P < 0.05). The mean histo-score of normal rims was intermediate between the two (P < 0.05). Cytoplasmic expression was strong in all autopsy-derived tissues, weak/negative in hyperplasia (100%), moderate in 16% adenomas, and 43% carcinomas.
Barazeghi et al. (2016) NR Uppsala, Sweden; St Leonards, Australia; London, UK 17 60 8 The epigenetic mark 5hmC is reduced in various cancers, and this may involve reduced expression of the TET1 enzyme. All 17 PC samples stained negatively for 5hmC. All 43 PA showed positive staining for 5hmC.
Bergero et al. (2005) 1974−2002 Turin, Italy 26 56 8 Gal-3 and Ki-67 were significantly higher in PC than PA, P < 0.05

The study suggests that Gal-3 immunostaining is a valuable tool to support a diagnosis of PC in highly proliferative tumors (Ki 67 > 6%) – sensitivity 96.2% and specificity 90%

Other markers tested: p27 and bcl2
Cavalier et al. (2014) NR Liège, Belgium 11 316 8 Automated 3rd/2nd generation PTH ratio was significantly higher in the PC group when compared to a control, P < 0.05. When elevated, 3rd/2nd generation PTH ratio had a sensitivity of 81.8% and specificity of 97.3% in the diagnosis of PC.
Ciregia et al. (2021) NR Pisa, Italy 6 11 6 Thirty-three differentially expressed proteins were identified in PC compared to PA. Among these, ubiquitin C-terminal hydrolase-L1 (UCH-L1) was highly overexpressed in PC.
Condello et al. (2021) NR Pisa, Italy 27 35 8 The expression of 740 genes are involved in the tumor progression process that was assessed in 8 PAs, 17 non-metastatic and 10 metastatic PCs. The comparison of gene expression between metastatic PCs and PAs revealed a signature of 103 genes. The comparison of gene expression between metastatic vs non-metastatic PCs showed the highest number of differentially expressed genes: 144. Genes that where associated with a deregulation in PC: CD24, FAP, FGFR1 and TBX1. Genes that were upregulated at the late stage of PC: MMP9, ANGPTL4, BMP7, FGFR1 and SOX2 genes and genes that were downregulated at the late stage of PC: ERBB3, TBX1, FBP1 and RAB25, possible contributing to the metastatic phenotype of PC through EMT and angiogenesis processes.
Cui et al. (2019) NR Beijing, China 19 19 8 19 samples of recurrent or metastatic PC

9/19 (47%) of PC samples harboured at least one potentially actionable genomic alteration

CDC73 mutations were detected in 9/19 (47%) PC
Erovic et al. (2012) 1998−2010 Toronto, Canada 10 35 7 Bcl-2a, parafibromin, Rb, and p27 were significantly decreased to variable degrees in all PC

Other markers tested: PDGFR-α, PDGFR-β, VEGFR-2, EGFR, COX-1, COX-2, MMP-1, CD9, keratin 7, Cdc2p34, cyclin D1, Rb, p27, p21, parafibromin, Bmi-1, 14-3-3σ, p53, Bcl-2a, Mcl-1, Bcl-xL, Gst-π, Smo, SHH, Gli-1, Gli-2, Gli-3, AKT, mTOR, FoxO-1, Wisp-1, Wisp-2
Guarnieri et al. (2012) 1990−2007 Foggia, Italy 15 46 9 CDC73 mutations were detected in 9/15 (60%) PC, 2/14 (14 %) AA, and 1/17 (6%) PA

Absence of parafibromin staining in 8/12 (67%) PC, 2/13 (15%) AA, and 3/17 (18%) PA
Hu et al. (2016) NR Beijing, China 12 37 8 The variations in mtDNA of tumor tissues and matched peripheral WBCs (white blood cells) in 12 PCs and 25 PAs were explored using ultradeep next-generation sequencing. A total of 821 germline variants and 23 somatic mutations were identified. Tumor somatic mutation in a coding sequence (CDS) with heteroplasmic factor (HF) >0.8 showed a higher mitochondrial DNA (mtDNA) copy number (P = 0.039). Tumor mtDNA copy number revealed a positive correlation with serum levels of intact parathyroid hormone and calcium. The results demonstrate that null recurrent mtDNA mutational hotspots were PC specific. An increased mtDNA copy number in parathyroid neoplasms was associated with somatic CDS mutations with a high HF and major clinical features.
Juhlin et al. (2007) NR Sweden (worldwide samples) 33 33 8 68% (15 out of 22) of the unequivocal carcinomas exhibited reduced expression of parafibromin while the 25 sporadic adenomas used as controls were entirely positive for parafibromin expression. Additionally, three out of the six carcinomas with known HRPT2 mutations showed reduced expression of parafibromin. Using all four antibodies, comparable results were obtained on the cellular level in individual tumors suggesting that there exists no epitope of choice in parafibromin immunohistochemistry. The results agree with the demonstration of a ~60 kDa product preferentially in the nuclear fraction by western blot analysis. Parafibromin immunohistochemistry could be used as an additional marker for parathyroid tumor classification, where positive samples have low risk of malignancy, whereas samples with reduced expression could be either carcinomas or rare cases of adenomas likely carrying an HRPT2 mutation.
Krupinova et al. (2021) NR Moscow, Russia 13 24 8 Significant changes in expression levels between the PC group (n=13) and the PA group (n=11) (P < 0.05) were observed for 17 microRNAs (miRNAs). Among them, the downregulation of miRNA-342-3p met the Benjamini-Hochberg adjustment criteria for multiple comparisons (P = 0.02).
Kumari et al. (2016) 1993−2013 Lucknow, India 14 227 8 Combination of PF, Gal-3 and PGP9.5 showed 50 % sensitivity, 97.9 % specificity and 95.4 % predictive accuracy for PC. Histological criteria still remains the most established method for predicting risk of malignancy in parathyroid neoplasms irrespective of whether old or revised criteria are used. Combination of positive (Gal-3, PGP9.5) and negative (PF) immunohistochemical markers may be used as an adjunct to histology in histological, atypical and malignant parathyroid neoplasms to obviate the need for repeated follow-up.
Kutahyalioglu et al. (2019) 2014−2016 Houston, USA 11 11 8 The most common mutations identified were in the PI3 K (PIK3CA, TSC1 and ATM) (4/11 patients) and TP53 (3/11) pathways. Genes not previously reported to be mutated in PC included: SDHA, TERT promoter and DICER1. Actionable mutations were found in 54% (6/11) of the patients.

Mutational profiling using NGS panels in advanced PC has yielded important potentially targetable genetic alterations.
Pandya et al. (2017) NR NR 17 17 9 CDC73 mutations were detected in 8/17 (47%) PC samples

New recurrent mutation found in putative kinase ADCK1
Rubin et al. (2008) NR New York, USA 8 18 9 The normal range for serum malignant hCG is not defined. In the benign PHPT subjects, the range of serum malignant hCG was 1.76–3.77 pmol/l. Serum malignant hyperglycosylated hCG values in all patients with PC exceeded the maximal serum malignant hCG level of the PHPT subjects with benign disease (3.77 pmol/l).
Silva-Figueroa et al. (2018) 1996−2006 Houston, USA 17 30 9 PC tends to have immune-ignorant and immune-tolerant microenvironments within the neoplasm (immunotypes II and IV). Of the PC microenvironments, 17 had patterns of programmed death-ligand 1 and tumor-infiltrating lymphocytes expression (immunotype I), suggesting possible benefit from immunotherapy. In addition, both PCs and parathyroid neoplasms expressed CD68 +.

No significant difference in the median programmed death-ligand 1 H score between PC and BPA

PCA had a lesser median CD3 + density than BPA (P = 0.04)

The study showed 4 out of 17 PCA patients had tumor microenvironments possibly suitable for anti-PD-L1 therapy (immunotype 1), 7 out of 18 PCA patients had a tumor microenvironment that would be possibly suitable for anti-CTL4 or anti-PD-1 therapy (immunotype 2).
Storvall et al. (2021) 2000−2011 Helsinki, Finland 32 151 8 PCs were characterized by increased cytoplasmic Filamin A expression compared to AA and PA, (P  = 0.004). Cytoplasmic Filamin A expression was also associated with higher serum calcium, PTH concentrations and male sex (P  = 0.014, P  = 0.017 and P  = 0.049 respectively). Parathyroid tumors with low Filamin A expression and positive parafibromin staining were extremely likely to be benign (P < 0.001).

A combined Filamin A and parafibromin expression score shows potential as a prognostic predictor of indolent behaviour in parathyroid neoplasms.
Uljanovs et al. (2021) NR Riga, Latvia 5 179 8 Proliferative lesions (PPH, adenoma and carcinoma) showed statistically significantly up-regulated CK19 (P = 0.012), decreased E-cadherin levels and distinctive patterns of vimentin. CD44, CD56 and p53 were almost absent from parathyroid tissues. All carcinomas lacked parafibromin contrasting with invariable positivity in adenomas.
Wang et al. (2012) 1992−2005 Beijing, China 15 42 7 Absence of parafibromin staining in 9/15 (60 %) PC and 1/26 (4%) PA, P < 0.001

Gal-3 staining was positive in 11/15 (73%) PC and 6/27 (22%) PA, P = 0.002

Ki-67 index was high (>5%) in 4/15 (27%) PC, 1/27 (4%) PA, P = 0.047

Combined specificity of positive Gal-3 and Ki-67 could reach 100%

Other markers tested: fragile histidine triad
Wang et al. (2021) NR Beijing, China 12 25 8 Multiple differentially expressed miRNAs of serum exosomes were screened out by sequencing. The expression of exosomal hsa-miR-27a-5p was upregulated in serum exosomes of PC patients, being able to clearly distinguish PC patients from PA controls
Witteveen et al. (2011) 1985−2000 Several hospitals, The Netherlands 23 23 7 Downregulation of CASR expression was found in 7 (30%) and associated with a 16x fold increase of developing metastasis, P < 0.05

Loss of parafibromin was found in 13 (59%) and associated with a 4-fold increased risk of metastasis, P < 0.05

CDC73 mutations were found in 4 (17%) and associated with a 7-fold increase of developing metastasis, P < 0.05

As bone manifestations were the most frequent symptoms/diseases at clinical presentation, Chen et al. (2003) analysed age-, gender- and race-adjusted bone mineral density (BMD), measured by dual-energy X-ray absorptiometry (DEXA) in 131 patients with PHPT, of which 20 patients had PC. They reported that the BMD of the distal one-third of the radius was significantly decreased in the PC group, compared to the BPA group. No reduction in BMD occurred in the lumbar spine, suggesting that osteopenia was worse in areas rich in cortical bone for PC patients (Chen et al. 2003).

PC presents equally in males and females with a median age of presentation of 44–65 years. Clinical symptoms are listed according to their prevalence: bone manifestation > renal manifestation > fatigue > palpable neck mass > neuropsychiatric symptoms.

Laboratory findings

PC accounts for only 12% of all cases of PHPT. The main laboratory findings in PC are a markedly raised corrected calcium and PTH levels Table 3. In a retrospective study of 131 consecutive patients with PHPT from Kobe University in Japan, 20 of whom had PC, serum levels of corrected calcium, alkaline phosphatase (ALP) and PTH were significantly higher in the malignant group when compared to the benign group (Chen et al. 2003). These biochemical findings are similar to those reported by Bae et al. (2012) in a cohort study from Seoul, South Korea, who showed an 83% sensitivity and 97% specificity for PC if ALP levels are greater than 285 IU/L. A nationwide study in Finland advises that PC should be suspected in any patient with PHPT and an ionised calcium concentration above 1.77 mmol/L (standard range, 1.151.30 mmol/L) (Ryhänen et al. 2017). In a Swiss study by Robert et al. (2005), which looked at 311 patients with PHPT, they found no patient with PC whose PTH value was under four-times the upper limit of normal. Absolute levels of PTH tend to be higher in PC than in BPA, but amino-PTH, an N-terminal-extended form of PTH, is overproduced in PC and very rarely seen in BPA (Cavalier et al. 2014). Cavalier et al. advocate using a third/second-generation PTH ratio which assesses for this over-production of amino-PTH, as only third generation PTH assays can measure amino-PTH. Values >1 of the third/second-generation PTH ratio have a sensitivity of 82% and a specificity of 97% as a marker for PC among PHPT patients. One study investigated human chorionic gonadotrophin (hCG) as a marker for PC and found that serum malignant hyperglycosylated hCG values, although without a quoted normal range, were higher in all eight patients with PC when compared to patients with PHPT (Rubin et al. 2008).

Absolute levels of PTH and ionised calcium concentrations tend to be significantly higher in PC than in BPA, and the additional use of a third/second-generation PTH ratio can help in the detection of PC.

Molecular pathogenesis

Multiple somatic gene defects have been found in PC samples, including mutations in CDC73, MEN1, CDKN1B, RET, PRUNE2, ADCK1, FAT3, AKAP9, ZEB1, SDHA, TERT promoter and DICER1 (Pandya et al. 2017, Cui et al. 2019, Kutahyalioglu et al. 2019). Mutations in cell division cycle-73 gene (CDC73, also known as HRPT2) were the most commonly implicated cause for PC and identified in 47–60% of PC samples (Guarnieri et al. 2012, Pandya et al. 2017, Cui et al. 2019). The CDC73 gene is a tumour suppressor gene located on chromosome 1q31.2 (Guarnieri et al. 2012). This gene encodes parafibromin, a protein involved in the regulation of gene expression and inhibition of cell proliferation. As a result, absence of parafibromin, caused by mutations in CDC73, makes parathyroid tissue more prone to carcinoma formation. PC most commonly occurs as a sporadic non-syndromic disorder. However, it has been associated with hereditary conditions that cause PHPT, such as multiple endocrine neoplasia type 1 and 2 (MEN1, MEN2), autosomal dominant familial isolated hyperparathyroidism (FIHP) and HPT-JT. HPT-JT is an autosomal dominant condition characterised by hyperparathyroidism secondary to multi-gland parathyroid neoplasia, and approximately 15% of patients with this syndrome have a risk of developing PC.

The calcium-sensing receptor (CASR) regulates synthesis and secretion of PTH and the proliferation of parathyroid glands (Witteveen et al. 2011). The CASR has been found to be downregulated in PC and also HPT-JT when compared to BPA (Witteveen et al. 2011). The study by Witteveen et al. (2011) of 23 patients with PC has shown CASR downregulation to be a negative prognostic indicator, with a reduced time to development of recurrence/metastasis and a lower 5-year disease-free survival. Condello et al. (2021) identified 144 genes that appeared to be associated with the transition from non-metastatic to metastatic PC. The researchers described the acquisition of additional genetic alterations at the late stage of PC, mainly addressing the upregulation of expression of MMP9, ANGPTL4, BMP7, FGFR1 and SOX2 genes and the downregulation of expression of ERBB3, TBX1, FBP1 and RAB25, being potential genes contributing to the metastatic phenotype of PC. Furthermore, the group confirmed the deregulation of genes already associated with PC, namely CD24, FAP, FGFR1 and TBX1.

PC most commonly occurs as a sporadic non-syndromic disorder but has been associated with hereditary conditions that cause PHPT, such as MEN1, MEN2, FIHP, especially HPT-JT. CDC73 mutations are the most commonly implicated cause for PC.

Imaging studies

Imaging investigation of parathyroid lesions has two different aims: (1) localisation of the parathyroid lesion prior to surgery and (2) distinction of benign from malignant parathyroid lesions.

Regarding the utility of imaging for localising PC prior to surgery, neck ultrasound (US) showed 71% sensitivity with a specificity of 100% (Harari et al. 2011). Other imaging modalities which may be considered are 4D-CT, 99mTC Sestamibi imaging SPECT/CT (MIBI), MRI and PET/CT with 2-[fluorine-18]-fluoro-2-deoxy-D-glucose (18F-FDG). Of these, 4-DCT and MIBI scanning are most commonly used. According to Christakis et al. (2017b), combined imaging with US, 4D-CT and MIBI has a 100% sensitivity for PC localisation, whereas imaging with only one modality has a poorer sensitivity (Christakis et al. 2017b , Lo et al. 2018). The use of MRI was recommended by one study as a tool to detect recurrence and metastasis, but data supporting this remain limited (Chen et al. 2018).

With respect to the distinction of benign from malignant parathyroid lesions, a tumour size of 30 mm (sensitivity 91%, specificity 92%) had a predictive value for the diagnosis of PC in patients with PHPT (Bae et al. 2012). Nonetheless, a US national database reported that one-third (33.8%) of PCs, in a population of 520 patients, had tumour sizes less than 30 mm (Lo et al. 2018). In a retrospective analysis of US features in parathyroid tumours larger than 15 mm, infiltration of surrounding tissue and calcification both had a 100% positive predictive value for malignant lesions, while high negative predictive values (NPV) were found for the absence of suspicious vascularity (NPV 97.6%), thick capsule (96.7%) and inhomogeneity (100%) (Sidhu et al. 2011). In relation to the the severity of the disease, Calapkulu et al. (2021) suggested tumour volume to be a more effective parameter than tumour size.

Combined imaging with US, 4D-CT and MIBI has a 100% sensitivity for PC localisation, whereas imaging with only one modality has a much poorer sensitivity. Infiltration of surrounding tissue and calcification both have a 100% positive predictive value for malignant lesions, while high negative predictive values (NPV) were found for the absence of suspicious vascularity (NPV 97.6%), thick capsule (96.7%) and inhomogeneity (100%).

Diagnosis and differential diagnosis

The differential diagnoses of raised PTH and a neck mass include PC and BPA as well as an atypical parathyroid neoplasm (APN). The management of these conditions vastly differs. Accurate assessment is therefore vital in the preoperative phase to ensure all PCs are managed adequately. Distinguishing between the three entities is challenging, as a diagnosis cannot be based solely on clinical, biochemical or imaging findings. Several studies have demonstrated that PC presents more aggressively with larger tumours (Bae et al. 2012), more severe hypercalcaemia (O’Neill et al. 2011, Bae et al. 2012, Quinn et al. 2015) and in younger patients than BPA (Chen et al. 2003, Karakas et al. 2012, Cavalier et al. 2014). Moreover, PC occurs equally between genders (Sadler et al. 2014), while BPA is more commonly found in women. Karakas et al. (2012) developed a logarithmic formula (Fig. 2) to calculate an individual’s risk of PC in those presenting with PHPT, based on preoperative serum calcium, PTH and age at diagnosis. They used their formula on 1256 patients presenting with PHPT, and by using a cut-off value of 5% for differentiation of the individual probabilities for the prediction of PC, identified all 19 patients who later were confirmed to have PC. The sensitivity of their algorithm was 100% and the specificity was 98%. While fine-needle aspiration and cytology were not recommended by Talat & Schulte (2010) due to fears of disrupting the tumour capsule and increasing the possibility of tumour implantation, Steen et al. found cytological differences between BPA and PC, with only PC showing pleomorphism with irregular and prominent nucleoli.

Figure 2
Figure 2

Logarithmic equation calculating preoperative risk of parathyroid cancer in patients with primary hyperparathyroidism by Karakas et al. (2012). A full colour version of this figure is available at https://doi.org/10.1530/ERC-22-0287.

Citation: Endocrine-Related Cancer 30, 4; 10.1530/ERC-22-0287

With regard to APN, the diagnosis remains a clinical challenge. APN has characteristics both of BPA and PC and is made through histology when parathyroid neoplasms have cytological features of PC but lack the defining characteristic of invasive growth (Christakis et al. 2016a ). Christakis et al. (2016a) compared 54 patients with a histopathological diagnosis of PC or APN in a retrospective review and found no significant group differences when comparing the clinical symptoms, whereas the PC group presented with significant higher PTH levels, with a 5-year disease-free survival rate for PC of 59.6%vs 90.9% for APN. Patients with tumour recurrence in the APN group presented soft tissue invasion in the initial histopathology report, but no other discrete features sufficient to be classified as PC. The authors concluded that a high index of clinical suspicion is needed preoperatively and intraoperatively to identify the parathyroid tumours that need oncological operations and recommend constant vigilance to identify the rare cases early that might behave in a malignant manner.

The differential diagnoses of raised PTH and a neck mass include PC and BPA as well as APN. PC presents more aggressively with larger tumours, more severe hypercalcaemia, and in younger patients than BPA, while BPA is more commonly found in women. The diagnosis of APN is made through histology and APN seems to have a benign course in the majority of cases, but long-term data are missing. Therefore, constant vigilance is required to identify the rare cases early that might behave in a malignant manner.

Pathology

The diagnosis of PC can only be confirmed with a detailed histopathology analysis (Talat & Schulte 2010). The World Health Organisation defines the presence of infiltrative growth (capsular and soft tissue invasion) or histological proof of PC vascular invasion as the minimum criteria for diagnosing PC (Lloyd et al. 2017). Fine-needle aspiration is not recommended as to avoid the incurable state of malignant parathyromatosis (Talat & Schulte 2010), even though Steen et al. found cytological characteristics (larger nuclei with more prominent atypia) that clearly set them apart from the adenomas. Histological features of PC include uniform sheets of cells arranged in a lobulated fashion, intervening fibrous trabeculae, capsule/vascular invasion (Sali et al. 2021) and the presence of mitotic figures. The presence of these findings has been assessed in an analysis of 1036 patients by Talat & Schulte (2010) who found that the presence of fibrous bands and the invasion of surrounding tissues are associated with lower recurrence and mortality rates at 5 years and overall better survival. In contrast, vascular invasion carries a 4-fold higher risk to experience death or recurrence rate at 5 years and a 2.8/2.6-fold higher risk of overall recurrence and death, respectively. Capsular invasion was the most reliable marker seen in PC, being present in 84% of cases, but has no prognostic impact (Talat & Schulte 2010). Liu et al. (2021) found that 57.1% (12/21) of patients with recurrent PC were diagnosed with benign parathyroid disease prior to the recurrence of PC, reflecting the considerable uncertainty regarding the pathological diagnosis of PC.

The histological diagnosis criteria of parathyroid carcinoma have changed between 2000 and 2020 with a number of papers reporting retrospective series of patients from the 1980s. Not all the studies included in this systematic review reported on criteria for PC diagnosis. Therefore, there may have been variability in the diagnostic criteria of studies included in this systematic review, another reason why the data were not pooled into a meta-analysis as stated previously.

Diagnosis of PC can only be confirmed with a detailed histopathology analysis with capsular invasion being the most reliable marker seen in PC, whereas fine needle aspiration is not recommended to avoid malignant parathyromatosis.

Immunohistochemistry and molecular analysis

Immunohistochemical markers are useful in the diagnosis of PC, and there are promising new markers that are potentially useful in the early diagnosis and prognosis of PC. Parafibromin, Ki-67 proliferation index and galectin-3 have the strongest evidence base for use in PC diagnosis (Erickson & Mete 2018). Absence of parafibromin has been assessed in multiple centres and identified in 45–68% patients with PC (Juhlin et al. 2007, Witteveen et al. 2011, Erovic et al. 2012, Guarnieri et al. 2012, Wang et al. 2012, Kumari et al. 2016). Parathyroid tumours with low filamin A expression and positive parafibromin staining, on the other hand, are extremely likely to be benign (P < 0.001), and this shows potential as a prognostic predictor of indolent behaviour in parathyroid neoplasms. The Ki-67 proliferation index may be helpful in diagnosing PC (Wang et al. 2012). Wang et al. (2012) showed that the Ki-67 proliferation index was high in 27% of PC compared to 5% of BPA. Bergero et al. (2005) also recommended its use following a study of 56 patients which showed that the Ki-67 proliferation index was higher in PC than BPA (6.7% vs 1.9%). Galectin-3 is recommended by Wang et al. (2012), who showed galectin-3 staining was positive in 73% of PC and 26% of BPA. Bergero et al. (2005) also recommended its use with their showing that galectin 3 levels were expressed by 24 of 26 of the PC patients (92.3%) but only in 1 (3.3%) out of 30 patients with BPA (P < 0.001). Additionally, all metastatic PCs (n = 6) were Gal-3–positive. Barazeghi et al. (2016) suggest 5-hydroxymethylcytosine (5hmC) as a novel epigenetic marker for the differentiation between PC and BPA, being negative in patients with PC and positive in patients with BPA.

Emerging evidence from Hu et al. (2022), Krupinova et al. (2021) and Wang et al. (2022), highlights the potential diagnostic role of microRNA- (miRNA) and microDNA- (miDNA) molecule expression in PC tumours. MiRNAs play an important role in the occurrence and metastasis of tumours as they are involved in almost all biological processes and affect cell proliferation, differentiation, adhesion, migration, invasion and apoptosis (Verdelli & Corbetta 2017). PC cancer can lead to alterations of circulating miRNA expression in the blood as reported by Krupinova et al. (2021). The group detected a downregulation of serum miRNA-342-3p in PC patients when compared to patients with BPA, suggesting miRNA-342-3p as promising biomarker for improvement of diagnosis of PC.

Wang et al. (2022) investigated miRNAs from serum exosomes of PC and BPA patients and found exosomal hsa-miR-27a-5p in serum exosomes of PC patients as a putative tumuor marker for preoperative identification of PC.

The combination of immunhistochemistry and molecular analysis can offer new perspectives for basic research and for future diagnostic and therapeutic strategies.

Treatment

Medical

Medical management is directed towards managing the effects of a raised PTH and subsequent hypercalcaemia. Intravenous fluids, diuretics and bisphosphonates are used to reduce serum calcium as first-line therapy. If first-line therapy is ineffective, the use of calcimimetic agents such as cinacalcet is recommended. Cinacalcet decreases PTH secretion and has been shown to be effective in the management of PC-induced hypercalcaemia, although more advanced tumours may noit express the CASR (Takeuchi et al. 2017).

Intravenous fluids, diuretics and bisphosphonates are used to reduce serum calcium as first-line therapy, whereas the use of calcimimetic agents such as cinacalcet is recommended as second-line therapy.

Surgical

Surgery is considered the gold standard of treatment for PC and should be performed at a reference centre, specialised in parathyroid surgery, to improve outcomes and provide the best chance of recovery (Kowalski et al. 2022). The type of surgery performed can be a localised excision following the parathyroid capsule as dissection plane or an oncological resection (also called ‘radical resection’ or ‘en bloc resection’) implying that this avoids capsule rupture, with removal of the parathyroid lesion with surrounding fat tissue, ipsilateral thyroid lobe and possibly the recurrent laryngeal nerve and ipsilateral central neck compartment lymph nodes, with a continuous margin of healthy tissue. Central locoregional lymph node metastasis may be present in around 10% of patients, while the lateral compartment is rarely involved (Schulte et al. 2010). There is significant debate as to which surgical technique should be utilised, as different studies have shown contradictory evidence. Schulte et al. (2014), in a study of 19 patients with PC, found that a primary oncological resection provides significantly better outcomes than a local excision, due to reduced R1 margins and therefore a lower chance of locoregional recurrence. Likewise, in a study of 38 patients, Iihara et al. (2007) found that those who underwent en bloc resections (22/38) had significantly longer disease-free survival in univariate analysis. This approach is supported by Iacobone et al. (2004), Kirkby-Bott et al. (2005), Mucci-Hennekinne et al. (2008), Karakas et al. (2012), Apaydin et al. (2021) and Xue et al. (2016), who recommend en bloc resection as the first-line option.

On the other hand, some papers support a less extensive surgical approach. (O’Neill et al. 2011, Harari et al. 2011, Young et al. 2016, Hu et al. 2019). For instance, O'Neill et al. (2011) attempted to address the question whether further radical surgery is required in patients diagnosed with PC following minimally invasive focused parathyroidectomy (MIP) and report on a small series of seven patients with diagnosis of PC after MIP. In this series, six out of seven patients underwent revision en bloc surgery which did not reveal any residual disease, and after a median follow-up of 44 months, there was no clear evidence for recurrence in any of these patients. The authors argue that in such selected patients, there might be no need for early completion surgery after PC diagnosis and also note that long-term data are missing. The authors state that they report on a subgroup of patients with PC, with disease localised to a single site, unlikely to have widespread local involvement, metastatic local nodal disease or distant metastases.

The largest study (n = 555) examining the impact of the extent of surgical resection on survival for PC was conducted by Leonard-Murali et al. (2021). The group found no significant difference in overall survival between patients with PC treated with local resection (n = 522) when compared to radical surgery (n = 33). They propose that patients who are found to have parathyroid carcinoma after undergoing local resection for a presumed benign indication may be safely and closely monitored for recurrence, rather than be committed to the morbidity of radical surgery. The study holds several limitations: first, Leonard-Murali et al. (2021) report only overall survival (OS) instead of progression-free survival or disease-specific survival as suitable parameter to advice for or against the surgical approach; and secondly, the groups in this study were stratified according to the operation code providing sufficient detail to distinguish local resection from radical surgery, but further information regarding extent of lymphadenectomy, specific adjacent organs removed and dissection planes was not available. Another interesting intraoperative approach was presented by Kowalski et al. (2022), who suggested intraoperative inspection of all four parathyroid glands if the diagnosis of PC is highly suspected during the primary operation.

From all available evidence, one must conclude that the gold standard for PC is still an en bloc resection. To ignore this, gold standard may lead to devastating outcomes for some of these patients (Iihara et al. 2007).

Although there is significant debate as to which surgical technique should be utilised, the gold standard for PC remains an en bloc resection.

Radiotherapy

There are no randomised controlled trials investigating the use of radiotherapy (RT) in PC and a clear consensus has not been reached regarding its use in patients with PC. RT has been used in the adjuvant setting for PC in a small number of patients (Busaidy et al. 2004, Erovic et al. 2013, Christakis et al. 2017a ). Christakis et al. (2017a) recommend its use, given its well-tolerated side-effects, in cases where there is concern for recurrence within the central compartment. They do not recommend the use of RT as treatment for metastatic disease, and the authors emphasised the need for clinicians to understand possible risks associated with RT, such as increased difficulty of subsequent neck surgery. Munson et al. (2003), Kirkby-Bott et al. (2005), Selvan et al. (2013) and Busaidy et al. (2004), also recommend post-operative RT, supporting its use in reducing local recurrence rates. Apaydin et al. (2021) suggest adjuvant radiotherapy to the neck to reduce the risk of local recurrence only in patients with microscopic residual parathyroid carcinoma.

However, the largest series from a single centre, although still only including just 11 patients, observed no significant effects on survival, stating that wide resections in initial surgical management are likely to be of greater importance (Erovic et al. 2013). Furthermore, two national registry analyses from the National Cancer Database, USA, independently showed that there was no improvement in overall survival rates in patients who received RT compared with patients who did not receive RT (Asare et al. 2015, Limberg et al. 2021). More importantly, Limberg et al. (2021) reported that 10.5% of patients with completely resected disease (M0, N0 or Nx) underwent radiotherapy without a benefit in overall survival (P = 0.183).

Currently, there are no randomised controlled trials investigating the use of RT in PC, and a clear consensus has not been reached regarding its use. Retrospective data show no improvement in OS rates in patients who received RT compared with patients who did not receive RT.

Chemotherapy and immunotherapy

Currently, chemotherapy (CT) and immunotherapy (IT) have not been shown to be effective in treating local or metastatic disease. Given that PC may express VEGFR-2, PDGFR-α and PDGFR-β mutations, Erovic et al. (2012) have suggested that sunitinib, bevacizumab and pazopanib, which are effective in the treatment of kidney, lung and breast malignancies, may be useful in PC. However, there are no clinical trials to support this. Silva-Figueroa et al. (2018) performed a retrospective study on 17 PC samples assessing the tumour microenvironment. The study showed that the majority of PC is PD-L1 negative, while three (17.6%) exhibited an adaptive immunotype suggesting suitability for anti-PD-L1 therapy (immunotype 1). The study advocates for more research into immune targeting in PC.

Currently, CT and IT have not been shown to be effective in treating local or metastatic disease.

Follow-up

The natural course of the disease is generally slow and inconsistenly progressive, with the eventual development of local recurrence and distant metastases. Recurrence of PC occurs in about 50% of cases (Talat & Schulte 2010) with a median disease-free interval after the initial surgery of 36 months (Liu et al. 2021), but longer intervals of 15–20 years have been reported (Iacobone et al. 2004). Given that PC can reoccur after 20 years, many centres recommend lifetime follow-up (Christakis et al. 2016b , Medas et al. 2016). In most instances, hypercalcaemia is the initial sign of recurrence (Busaidy et al. 2004). Liu et al. (2021) evaluated the sonographic findings of locoregional recurrences of PC and found hypoechoic solid nodules in 28 relapses (96.6%), inhomogeneous echotexture in 28 relapses (96.6%) and intralesional echogenic septa-like structures in 21 relapses (72.4%), as well as irregular shape in 22 relapses (75.9%), marked vascularisation on colour Doppler imaging in 19 relapses (65.5%) and multiple lesions in 26 relapses (89.7%). None of the recurrent lesions exhibited calcification, and the total size of all the recurrent lesions at each relapse mildly correlated with the serum intact parathyroid hormone levels (r = 0.450; P = 0.014). Distant metastases most frequently occur within the lungs (Hu et al. 2019) and bones (Iacabone et al. 2004). As noted above, currently, there is no evidence supporting the routine use of chemotherapy, immunotherapy or radiotherapy in the metastatic setting. Once PC is metastatic, cure is highly unlikely and most patients die from severe hypercalcaemia or metabolic complications rather than metastases per se.

Recurrence of PC occurs in about 50% and lifetime follow-up ist recommended with hypercalcaemia being the initial sign of recurrence. Once PC ist metastatic, cure is highly unlikely.

Survivorship and prognostic indicators

Five-year overall survival (OS) ranges from 60% to 93% (Busaidy et al. 2004, Kleinpeter et al. 2005, Karakas et al. 2012, Hsu et al. 2014, Christakis et al. 2016b , Young et al. 2016, Ryhänen et al. 2017, Kong et al. 2021, Wei et al. 2022). Combined assessment of prognostic factors is challenging as many studies evaluated different outcomes such as 5-year OS, 10-year OS, disease-free survival (DFS) and disease-specific survival (DSS). Greater age has been found to be a negative prognostic factor by Sadler et al. (2014), Hu et al. (2019) and Kong et al. (2021) but a positive prognostic factor by Talat & Schulte (2010) and non-significant by Harari et al. (2011). In the largest study by Sadler et al. (2014), mean OS was lower in patients who were Black, older than 57 years of age, had more than two comorbidities, and whose tumour was a secondary malignancy. Additionally, positive surgical margins, positive lymph nodes and poorly differentiated histology all conferred a worse OS. However, in a review of 330 patients by Talat & Schulte (2010), differing results were found, with male gender, younger age and high calcium being adverse clinical prognostic factors. They agreed that positive surgical margins carried a poorer prognosis. An analysis of the SEER registry between 1973 and 2014 revealed that patients with distant metastasis, but not metastatic lymph nodes, and PC tumours >30 mm had worse DSS (Lo et al. 2018). Lenschow et al. (2022) reported that by TNM a low T status,N0 stage at initial diagnosis and post-operative biochemical remission were beneficial prognostic parameters for recurrence-free survival of PC. The type of surgical procedure to treat PC has been debated heavily, and different centres still disagree over whether it affects prognosis (Kleinpeter et al. 2005, Young et al. 2016). A decrease in mortality and complication rate was observed when the initial surgery was conducted in an academic tertiary care endocrine surgery referral centre (Harari et al. 2011). With respect to revision surgery, Wei et al. (2022) and Wachter et al. (2019) found different results regarding recurrent or persistent PC, with (extended) en bloc resection (EEBR) providing better outcomes than other conventional surgical approaches such as parathyroidectomy only. Wei et al. (2022) reported a 5-year OS rate after EEBR of 59.6% compared to 16.7% after less radical procedures, with an improved median expected survival time of 90.0vs 13.0 months after local excision. Extended en bloc resection might therefore offer a second chance of cure for patients with recurrent or persistent PC in the absence of distant metastasis. Looking at immunhistochemical markers, a study by Hu et al. (2019) of 53 patients with PC showed that negative parafibromin correlated with a higher risk of recurrence and metastasis than in patients with positive parafibromin staining (Hu et al. 2019). The presence of a high Ki-67 proliferation index (>5%) has also been observed as a negative prognostic indicator in PC, as it was found to be associated with a higher chance of recurrence in a study of 38 patients by Iihara et al. (2007). Interestingly, clinicians at the M. D. Anderson Cancer Center (Christakis et al. 2016b ) compared the management of PC and outcomes (OS and DFS) in its centre between 1980–2001 and 2002–2015, and no significant changes in OS and DFS were observed over 35 years, highlighting the need to improve oncologic care.

Prognostic indicators vary between studies, but positive surgical margins, a greater age, the presence of a high Ki-67 proliferation index as well as negative parafibromin staining, seem to carry a poorer prognosis. Extended en bloc resection, in an academic tertiary care endocrine surgery referral centre, is the primary mode of therapy for recurrence of PC. However, reoperation is rarely curative and eventual relapse is likely.

Prognostic tools and staging

Three prognostic tools have been proposed in the management of PC, one by Shaha & Shah (1999) and two by Talat & Schulte (2010). The validity of Shaha & Shah’s prognostic tool has been reviewed by Talat & Schulte (2010), who found no significant difference overall between the groups when they used this tool to assess 185 patients with PC. Talat & Schulte (2010)proposed 2 new prognostic tools which are based on anatomical disease progression: a high and low risk stratifying tool and a stage I to IV based on the TNM scale. Use of these Schulte prognostic tools has been tested by Sadler et al. (2014) using 91 patients with PC from a database of 1022 patients with PC, and they found that the high/low-risk tool was an effective method of predicting better outcomes with a 28-month greater mean survival in the low-risk group. Villar-del Moral et al. (2014) also support the use of the Schulte tool after their study of 62 patients with PC in Spain showed a significant difference in tumour recurrence in Stage III PC patients when compared to Stage I. Furthermore, they state that the Shaha & Shah system was inferior to Schulte’s, as it did not show a stepwise worsening of prognosis at greater stages in the prognostic tool, unlike the Schulte tool. Likewise, Xue et al. (2016) advocate for the use of Schulte’s 4 stage classification system and high/low-risk classification for recurrence, finding a significant difference for recurrence in 40 patients with PC in China. Schulte et al. (2012) have also validated the use of their prognostic classification system in their review of 82 patients. Their study showed a significant impairment in survival (98.6% I, 79.2% II, 71.4% III, 40.0% IV, P < 0.05 between each class). The American Joint Committee on Cancer (AJCC) eighth edition of cancer staging has proposed a classification of PC in 2017.

We have created an updated staging system based on the eighth edition of the American Joint Committee on Cancer (AJCC) TNM staging (Supplementary Table 3).

Discussion

Based on current evidence, we have proposed a diagnostic algorithm for patients presenting with PTHP (Fig. 3).

Figure 3
Figure 3

Diagnostic algorithm for parathyroid carcinoma. A full colour version of this figure is available at https://doi.org/10.1530/ERC-22-0287.

Citation: Endocrine-Related Cancer 30, 4; 10.1530/ERC-22-0287

PC should be suspected over other causes of PHPT whenever patients have a personal or family history of HPT-JT, exhibit markedly elevated calcium and PTH levels, or present with severe symptoms. However, anecdoctal cases of normocalcaemic hyperparathyroidism have also been reported (Al-Kurd et al. 2014). We recommend that all patients undergo an initial US scan of the neck. The possible parathyroid lesion should then be classified as suspicious or non-suspicious. Thereafter, patients who are severely symptomatic, have a personal or family history of HPT-JT, or have a suspicious lesion on the US, should undergo further investigations including a screening for the most common manifestations involving the bones and kidneys.

If a lesion is classified as non-suspicious, laboratory results must be reviewed and decisions on further management should be taken in a multidisciplinary manner. Unlike Schulte et al. (2012) in their diagnostic algorithm, we do not suggest calcium and tumour size cut-offs, given the increasing evidence that PC is also present in smaller tumours and in patients who do not present with severe hypercalcaemia. For further investigations, we recommend the use of a second-/third-generation PTH assay ratio and imaging with 4-DCT and MIBI. The use of the tool created by Karakas et al. requires validation, but it could be a useful diagnostic adjunct in the preoperative phase. If the second-/third- generation PTH assay ratio and imaging with 4-DCT and MIBI are negative, follow-up as a likely benign PTHP is proposed. If any are positive, we recommend that the patient undergoes an oncological en bloc resection with curative intent. Although the extent of surgical resection required to ensure optimal clinical outcomes is not entirely clear, we advise en bloc resection as gold standard for the surgical approach due to better outcomes with regard to local disease control. The diagnosis of APN, which is an important differential diagnosis of PC, is made through histology. APN seems to have a benign course in the majority of cases but long-term data are missing. Therefore, constant vigilance is required to identify the rare cases early that might behave in a malignant manner.

In case of PC diagnosis, samples should be sent for genetic and immunohistochemical analysis post-operatively. We recommend that the genetic analysis should be concerned with defects in CDC73, and immunohistochemical analysis with parafibromin, Ki-67 proliferation index and galectin-3. Cardoso et al. (2017) performed a comprehensive review on the molecular genetics of PC. Their findings, like others, suggest that CDC73 mutations are major drivers in the aetiology of PC. They go on to suggest that PC involves other genes such as MEN1, RET and PRUNE2, as well as epigenetic mechanisms, alterations in miRNA expression and potentially other, as yet unidentified, genes. In patients with MEN-1, parathyroid tumours are rarely PC (Cardoso et al. 2017). With respect to the diagnostic performance of parafibromin, Hu et al. (2016) completed a meta-analysis of ten studies. Their study showed that the pooled specificity of parafibromin staining was satisfactory for the diagnosis of PC (95% CI: 85–98%), while the sensitivity was limited 68% (95% CI: 49–82%). This is in line with the findings of Uljanovs et al. (2021). The aim of this study was to assess the molecular landscape and its heterogeneity in primary parathyroid hyperplasia (PPH) and adenoma, compared to carcinoma and normal glands. All carcinomas lacked parafibromin contrasting with invariable positivity in adenomas. Remarkable heterogeneity of cell cycle markers and intermediate filaments must be accounted for in scientific studies and elaboration of diagnostic cut-offs. Given the challenges in characterising PC, a combination of markers may be required to achieve a definitive diagnosis.

With regard to the use of chemotherapy, Adam et al. (2010) assessed in a review trial of doxorubicin, cyclophosphamide and 5-flurouracil, and dacarbazine, cyclophosphamide, 5-fluorouracil, and vincristine, which deemed to be ineffective in the management of PC. Given the poor evidence-base supporting use of chemotherapy and immunotherapy, we would currently not recommend their routine use in the management of PC. To date, there are no reports that have shown any survival benefit of radiotherapy in the treatment of PC (Asare et al. 2015, Limberg et al. 2021). Adjuvant radiotherapy is therefore not recommended as standard therapy for PC and there is a certain risk of overutilisation, without a benefit in OS, particularly in patients with completely resected localised disease undergoing radiotherapy, as reported by Limberg et al. (2021). Unlike other endocrine cancers, there is no established TNM classification, due to lack of data. Overall, the evidence supports the use of both of Schulte’s criteria for the prognosis of PC to assist in clinical judgement. In terms of follow-up, Schulte & Talat (2012) delineated different follow-up plans depending on whether the patient was classified as high or low risk after the initial surgery. Low-risk patients should have circulating PTH and calcium checked at 6-month intervals for the first 5 years, and annually thereafter, patients with high-risk disease require closer follow-up with 3 monthly reviews for the first decade and 6 monthly reviews thereafter. If PTH or calcium is elevated, it is recommended that patients undergo localising studies such as 4D-CT and MIBI scanning and PTH assay ratio measurements for the assessment of recurrent disease. When PC is no longer amenable to surgical resection, managing hypercalcaemia can prolong survival (Ramos et al. 2017). Guise & Wysolmerski (2022)suggested in their review Cancer-Associated Hypercalcaemia, the correction of volume depletion and the inhibiton of bone resorption are the two basic principles of refractory hypercalcaemia next to the initial treatment of the underlying cancer. Antiresorptive agents include bisphosphonates, calcitonin and denosumab. In particular, denosumab, as a human MAB to the receptor activator of NF-κB ligand (RANKL), although not approved for this indication, has been successfully used in PC patients with refractory hypercalcaemia (Bowyer et al. 2013, Karuppiah et al. 2014, Vellanki et al. 2014), leading to an improvement of symptoms and quality of life (Roukain et al. 2021). Cinacalcet, on the other hand, as an oral calcimimetic agent that reduces PTH secretion and blocks renal tubular reabsorption of calcium, is approved for the treatment of PC-associated hypercalcaemia and reduces calcium levels in approximately 60% of patients with PC (Silverberg et al. 2007, Guise & Wysolmerski 2022). Case reports have described its safety and effectiveness in treating several cases of malignancy-associated hypercalcemia alone (Silverberg et al. 2007) or in combination with other medications (Tzotzas et al. 2022), and even throughout pregnancy (Horjus et al. 2009, Nadarasa et al. 2014).

The limitations of this review are due to the significant heterogeneity and retrospective nature of studies, and study design largely limited to single-centre experiences with a reduced sample size. Another limitation is the fact that not all the studies included into this systematic review report on criteria for PC diagnosis. Therefore, there may have been variability in the diagnostic criteria of studies included in this systematic review, which range between 1980 and 2022.

We believe that the study of rare diseases, such as PC, would benefit from multicentre collaborations with standardised databases (Clayman et al. 2004). Furthermore, patients with PC would benefit from inclusion in clinical trials of new targeted agents in which the mutation, regardless of the origin of the tumour, is the criteria for eligibility.

Conclusions

PC is a rare disease which typically presents with hypercalcaemia and often with systemic symptoms. Early recognition of lesions at risk of being PC supported by genetic, biochemistry and imaging is required to determine the surgical approach and extent of resection, with en bloc resection remaining the gold standard for surgical approach. Diagnosis is generally confirmed post-operatively by immunohistochemical and pathological analysis. Given the lack of alternative curative approaches and limited outcome benefit from revisional surgery, we recommend referral to expert centres with experience in oncological parathyroid surgery. However, more evidence is required to ascertain the adequate surgical approach and role of radiotherapy as standards of care. Emerging evidence indicates that targeted therapy based on molecular biomarkers may be a novel treatment option. The rarity of PC and need for personalised treatment warrant multidisciplinary management in an experienced centre.

Supplementary materials

This is linked to the online version of the paper at https://doi.org/10.1530/ERC-22-0287.

Declaration of interest

The authors declare that they have no conflict of interest.

Funding

National Institute for Health Research (NIHR), South London Clinical Research Network (CRN) ‘Green shoots’ Investigator Award supported Dr Dimitriadis in the writing of this manuscript.

References

  • Adam MA, Untch BR & Olson JA 2010 Parathyroid carcinoma: current understanding and new insights into gene expression and intraoperative parathyroid hormone kinetics. Oncologist 15 6172. (https://doi.org/10.1634/theoncologist.2009-0185)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Agarwal S, Kardam S, Chatterjee P, Kumar C, Boruah M, Sharma MC, Tabin M & Ramakrishnan L 2022 CaSR expression in normal parathyroid and PHPT: new insights into pathogenesis from an autopsy-based study. Journal of Endocrinological Investigation 45 337346. (https://doi.org/10.1007/s40618-021-01646-w)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Al-Kurd A, Mekel M & Mazeh H 2014 Parathyroid carcinoma. Surgical Oncology 23 107114. (https://doi.org/10.1016/j.suronc.2014.03.005)

  • Apaydın T & & Yavuz DG 2021 Seven cases of parathyroid carcinoma and review of the literature. Hormones (Athens) 20 189195. (https://doi.org/10.1007/s42000-020-00220-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Asare EA, Sturgeon C, Winchester DJ, Liu L, Palis B, Perrier ND, Evans DB, Winchester DP & Wang TS 2015 Parathyroid carcinoma: an update on treatment outcomes and prognostic factors from the national cancer data base (NCDB). Annals of Surgical Oncology 22 39903995. (https://doi.org/10.1245/s10434-015-4672-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bae JH, Choi HJ, Lee Y, Moon MK, Park YJ, Shin CS, Park DJ, Jang HC, Kim SY & Kim SW 2012 Preoperative predictive factors for parathyroid carcinoma in patients with primary hyperparathyroidism. Journal of Korean Medical Science 27 890895. (https://doi.org/10.3346/jkms.2012.27.8.890)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barazeghi E, Gill AJ, Sidhu S, Norlén O, Dina R, Palazzo FF, Hellman P, Stålberg P & Westin G 2016 5-hydroxymethylcytosine discriminates between parathyroid adenoma and carcinoma. Clinical Epigenetics 8 31. (https://doi.org/10.1186/s13148-016-0197-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Basceken SI, Genc V, Ersoz S, Sevim Y, Celik SU & Bayram IK 2015 Is local resection sufficient for parathyroid carcinoma? Clinics (Sao Paulo) 70 247249. (https://doi.org/10.6061/clinics/2015(0405)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bergero N, De Pompa R, Sacerdote C, Gasparri G, Volante M, Bussolati G & Papotti M 2005 Galectin-3 expression in parathyroid carcinoma: immunohistochemical study of 26 cases. Human Pathology 36 908914. (https://doi.org/10.1016/j.humpath.2005.06.020)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bowyer SE, White AM, Ransom DT & Davidson JA 2013 Resistant hypercalcaemia in metastatic parathyroid carcinoma. Medical Journal of Australia 198 559561. (https://doi.org/10.5694/mja12.11243)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Busaidy NL, Jimenez C, Habra MA, Schultz PN, El‐Naggar AK, Clayman GL, Asper JA, Diaz EM, Evans DB & Gagel RF et al.2004 Parathyroid carcinoma: a 22-year experience. Head and Neck 26 716726. (https://doi.org/10.1002/hed.20049)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Çalapkulu M, Sencar ME, Unsal IO, Sakiz D, Duger H, Özbek M & & Çakal E 2021 Tumor volume can be used as a parameter indicating the severity of disease in parathyroid cancer. Endocrine Practice 27 706709. (https://doi.org/10.1016/j.eprac.2021.01.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cardoso L, Stevenson M & Thakker RV 2017 Molecular genetics of syndromic and non-syndromic forms of parathyroid carcinoma. Human Mutation 38 16211648. (https://doi.org/10.1002/humu.23337)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cavalier E, Betea D, Schleck ML, Gadisseur R, Vroonen L, Delanaye P, Daly AF & Beckers A 2014 The third/second generation PTH assay ratio as a marker for parathyroid carcinoma: evaluation using an automated platform. Journal of Clinical Endocrinology and Metabolism 99 E453E457. (https://doi.org/10.1210/jc.2013-3730)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen Q, Kaji H, Nomura R, Sowa H, Yamauchi M, Tsukamoto T, Yamaguchi T, Kobayashi A, Sugimoto T & Chihara K 2003 Trial to predict malignancy of affected parathyroid glands in primary hyperparathyroidism. Endocrine Journal 50 527534. (https://doi.org/10.1507/endocrj.50.527)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen Z, Fu J, Shao Q, Zhou B & Wang F 2018 99mTc-MIBI single photon emission computed tomography/computed tomography for the incidental detection of rare parathyroid carcinoma. Medicine 97 e12578. (https://doi.org/10.1097/MD.0000000000012578)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Christakis I, Bussaidy N, Clarke C, Kwatampora LJ, Warneke CL, Silva AM, Williams MD, Grubbs EG, Lee JE & Perrier ND 2016a Differentiating atypical parathyroid neoplasm from parathyroid cancer. Annals of Surgical Oncology 23 28892897. (https://doi.org/10.1245/s10434-016-5248-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Christakis I, Silva AM, Kwatampora LJ, Warneke CL, Clarke CN, Williams MD, Grubbs EG, Lee JE, Busaidy NL & Perrier ND 2016b Oncologic progress for the treatment of parathyroid carcinoma is needed. Journal of Surgical Oncology 114 708713. (https://doi.org/10.1002/jso.24407)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Christakis I, Silva AM, Williams MD, Garden A, Grubbs EG, Busaidy NL, Lee JE, Perrier ND & Zafereo M 2017a Postoperative local-regional radiation therapy in the treatment of parathyroid carcinoma: the MD Anderson experience of 35 years. Practical Radiation Oncology 7 e463e470. (https://doi.org/10.1016/j.prro.2017.05.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Christakis I, Vu T, Chuang HH, Fellman B, Figueroa AMS, Williams MD, Busaidy NL & Perrier ND 2017b The diagnostic accuracy of neck ultrasound, 4D-Computed tomographyand sestamibi imaging in parathyroid carcinoma. European Journal of Radiology 95 8288. (https://doi.org/10.1016/j.ejrad.2017.07.026)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ciregia F, Cetani F, Pardi E, Soggiu A, Piras C, Zallocco L, Borsari S, Ronci M, Caruso V, Marcocci C, Mazzoni MR, Lucacchini A, Giusti L. 2021 Parathyroid carcinoma and adenoma co-existing in one patient: case report and comparative proteomic analysis. Cancer Genomics Proteomics. Cancer Genomics Proteomics 18 781796. (https://doi.org/10.21873/cgp.20297)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clayman GL, Gonzalez HE, El‐Naggar A & Vassilopoulou‐Sellin R 2004 Parathyroid carcinoma: evaluation and interdisciplinary management. Cancer 100 900905. (https://doi.org/10.1002/cncr.20089)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Condello V, Cetani F, Denaro M, Torregrossa L, Pardi E, Piaggi P, Borsari S, Poma AM, Muscarella LA, Graziano P. 2021 Gene expression profile in metastatic and non-metastatic parathyroid carcinoma. Endocrine-Related Cancer 28 111134. (https://doi.org/10.1530/erc-20-0450)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cui M, Hu Y, Bi Y, Wang W, Wang M, Zhang X, Zhang R, Wang P, Su Z & Gao X et al.2019 Preliminary exploration of potential molecular therapeutic targets in recurrent and metastatic parathyroid carcinomas. International Journal of Cancer 144 525532. (https://doi.org/10.1002/ijc.31948)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • De Pasquale L, Bulfamante AM, Felisati G, Castellani L, Ghilardi G & Saibene AM 2021 Management and outcome of parathyroid carcinoma-induced primary hyperparathyroidism: a single-centre experience. International Journal of Endocrinology 2021 5397941. (https://doi.org/10.1155/2021/5397941)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Erickson LA & Mete O 2018 Immunohistochemistry in diagnostic parathyroid pathology. Endocrine Pathology 29 113129. (https://doi.org/10.1007/s12022-018-9527-6)

  • Erovic BM, Harris L, Jamali M, Goldstein DP, Irish JC, Asa SL & Mete O 2012 Biomarkers of parathyroid carcinoma. Endocrine Pathology 23 221231. (https://doi.org/10.1007/s12022-012-9222-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Erovic BM, Goldstein DP, Kim D, Mete O, Brierley J, Tsang R, Freeman JL, Asa SL, Rotstein L & Irish JC 2013 Parathyroid cancer: outcome analysis of 16 patients treated at the Princess Margaret Hospital. Head and Neck 35 3539. (https://doi.org/10.1002/hed.22908)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gill AJ 2014 Understanding the genetic basis of parathyroid carcinoma. Endocrine Pathology 25 3034. (https://doi.org/10.1007/s12022-013-9294-3)

  • Guarnieri V, Battista C, Muscarella LA, Bisceglia M, de Martino D, Baorda F, Maiello E, D’Agruma L, Chiodini I & Clemente C et al. 2012 CDC73 mutations and parafibromin immunohistochemistry in parathyroid tumors: clinical correlations in a single-centre patient cohort. Cellular Oncology 35 411422. (https://doi.org/10.1007/s13402-012-0100-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guise TA & Wysolmerski JJ 2022 Cancer-associated hypercalcemia. New England Journal of Medicine 386 14431451. (https://doi.org/10.1056/NEJMcp2113128)

  • Harari A, Waring A, Fernandez-Ranvier G, Hwang J, Suh I, Mitmaker E, Shen W, Gosnell J, Duh QY & Clark O 2011 Parathyroid carcinoma: a 43-year outcome and survival analysis. Journal of Clinical Endocrinology and Metabolism 96 36793686. (https://doi.org/10.1210/jc.2011-1571)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Horjus C, Groot I, Telting D, van Setten P, van Sorge A, Kovacs CS, Hermus A & de Boer H 2009 Cinacalcet for hyperparathyroidism in pregnancy and puerperium. Journal of Pediatric Endocrinology and Metabolism 22 741749. (https://doi.org/10.1515/jpem.2009.22.8.741)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hsu KT, Sippel RS, Chen H & Schneider DF 2014 Is central lymph node dissection necessary for parathyroid carcinoma? Surgery 156 13361341. (https://doi.org/10.1016/j.surg.2014.08.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hu Y, Liao Q, Cao S, Gao X & Zhao Y 2016 Diagnostic performance of parafibromin immunohistochemical staining for sporadic parathyroid carcinoma: a meta-analysis. Endocrine 54 612619. (https://doi.org/10.1007/s12020-016-0997-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hu Y, Bi Y, Cui M, Zhang X, Su Z, Wang My, Hua S, Liao Q & Zhao Y 2019 The influence of surgical extent and parafibromin staining on the outcome of parathyroid carcinoma: 20-year experience from a single institute. Endocrine Practice 25 634641. (https://doi.org/10.4158/EP-2018-0538)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hu Y, Zhang X, Wang O, Xing X, Cui M, Wang M, Song C & Liao Q 2021 Spectrum of mitochondrial genomic variation in parathyroid neoplasms. Endocrine. 74 690697. (https://doi.org/10.1007/s12020-021-02825-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hu Y, Cui M, Chang X, Wang O, Chen T, Xiao J, Wang M, Hua S & & Liao Q 2022 Patterns and predictors of cervical lymph node metastasis in parathyroid carcinoma. Cancers (Basel) 14 4004. (https://doi.org/10.3390/cancers14164004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iacobone M, Lumachi F & Favia G 2004 Up-to-date on parathyroid carcinoma: analysis of an experience of 19 cases. Journal of Surgical Oncology 88 223228. (https://doi.org/10.1002/jso.20152)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iihara M, Okamoto T, Suzuki R, Kawamata A, Nishikawa T, Kobayashi M & Obara T 2007 Functional parathyroid carcinoma: long-term treatment outcome and risk factor analysis. Surgery 142 936943.e1. (https://doi.org/10.1016/j.surg.2007.09.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Juhlin CC, Villablanca A, Sandelin K, Haglund F, Nordenström J, Forsberg L, Bränström R, Obara T, Arnold A & Larsson C et al.2007 Parafibromin immunoreactivity: its use as an additional diagnostic marker for parathyroid tumor classification. Endocrine-Related Cancer 14 501512. (https://doi.org/10.1677/ERC-07-0021)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Karakas E, Müller HH, Lyadov VK, Luz S, Schneider R, Rothmund M, Bartsch DK & Schlosser K 2012 Development of a formula to predict parathyroid carcinoma in patients with primary hyperparathyroidism. World Journal of Surgery 36 26052611. (https://doi.org/10.1007/s00268-012-1707-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Karuppiah D, Thanabalasingham G, Shine B, Wang LM, Sadler GP, Karavitaki N & Grossman AB 2014 Refractory hypercalcaemia secondary to parathyroid carcinoma: response to high-dose denosumab. European Journal of Endocrinology 171 K1K5. (https://doi.org/10.1530/EJE-14-0166)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kirkby-Bott J, Lewis P, Harmer CL & Smellie WJB 2005 One stage treatment of parathyroid cancer. European Journal of Surgical Oncology 31 7883. (https://doi.org/10.1016/j.ejso.2004.06.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kleinpeter KP, Lovato JF, Clark PB, Wooldridge T, Norman ES, Bergman S & Perrier ND 2005 Is parathyroid carcinoma indeed a lethal disease? Annals of Surgical Oncology 12 260266. (https://doi.org/10.1245/ASO.2005.03.036)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kong SH, Kim JH, Park MY, Kim SW & & Shin CS 2021 Epidemiology and prognosis of parathyroid carcinoma: real-world data using nationwide cohort. Journal of Cancer Research and Clinical Oncology 147 30913097. (https://doi.org/10.1007/s00432-021-03576-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kowalski GJ, Bednarczyk A, Buła G, Gawrychowska A & Gawrychowski J 2022 Parathyroid carcinoma - a study of 29 cases. Endokrynologia Polska 73 5663. (https://doi.org/10.5603/EP.a2022.0003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kumari N, Chaudhary N, Pradhan R, Agarwal A & Krishnani N 2016 Role of histological criteria and immunohistochemical markers in predicting risk of malignancy in parathyroid neoplasms. Endocrine Pathology 27 8796. (https://doi.org/10.1007/s12022-016-9426-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kutahyalioglu M, Nguyen HT, Kwatampora L, Clarke C, Silva A, Ibrahim E, Waguespack SG, Cabanillas ME, Jimenez C & Hu MI et al.2019 Genetic profiling as a clinical tool in advanced parathyroid carcinoma. Journal of Cancer Research and Clinical Oncology 145 19771986. (https://doi.org/10.1007/s00432-019-02945-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Krupinova J, Mokrysheva N, Petrov V, Pigarova E, Eremkina A, Dobreva E, Ajnetdinova A, Melnichenko G & & Tiulpakov A 2021 Serum circulating miRNA-342-3p as a potential diagnostic biomarker in parathyroid carcinomas: a pilot study. Endocrinology, Diabetes & Metabolism 4 e00284. (https://doi.org/10.1002/edm2.284)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee PK, Jarosek SL, Virnig BA, Evasovich M & Tuttle TM 2007 Trends in the incidence and treatment of parathyroid cancer in the United States. Cancer 109 1736–1741. (https://doi.org/10.1002/cncr.22599)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee YS, Hong SW, Jeong JJ, Nam KH, Chung WY, Chang HS & Park CS 2010 Parathyroid carcinoma: a 16-year experience in a single institution. Endocrine Journal 57 493497. (https://doi.org/10.1507/endocrj.k09e-365)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lenschow C, Schrägle S, Kircher S, Lorenz K, Machens A, Dralle H, Riss P, Scheuba C, Pfestroff A & Spitzweg C et al.2022 Clinical presentation, treatment, and outcome of parathyroid carcinoma: results of the NEKAR retrospective international multicenter study. Annals of Surgery 275 e479e487. (https://doi.org/10.1097/SLA.0000000000004144)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leonard-Murali S, Ivanics T, Kwon DS, Han X, Steffes CP & & Shah R 2021 Local resection versus radical surgery for parathyroid carcinoma: a National Cancer Database analysis. European Journal of Surgical Oncology 11 27682773. (https://doi.org/10.1016/j.ejso.2021.06.026)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Limberg J, Stefanova D, Ullmann TM, Thiesmeyer JW, Bains S, Beninato T, Zarnegar R, Fahey 3rd TJ & & Finnerty BM 2021 The use and benefit of adjuvant radiotherapy in parathyroid carcinoma: a National Cancer Database analysis. Annals of Surgical Oncology 28 502511. (https://doi.org/10.1245/s10434-020-08825-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu Y, Li J, Liu H, Yang H, Qiao J, Tao Wei 4, Wang T & & Yu Y 2021 Spontaneous remission after a hypercalcemic crisis caused by an intracystic hemorrhage of bilateral parathyroid adenomas: a case report and literature review. Frontiers in Endocrinology (Lausanne) 12 766234. (https://doi.org/10.3389/fendo.2021.766234)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lloyd RV, Osamura RY, Klöppel G & Rosai J 2017 WHO classification of tumours of endocrine organs. International Agency for Research on Cancer. (available at https://publications.iarc.fr/Book-And-Report- Series/Who-Classification-Of-Tumours/WHO-Classification-Of- Tumours-Of-Endocrine-Organs-2017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lo WM, Good ML, Nilubol N, Perrier ND & Patel DT 2018 Tumor size and presence of metastatic disease at diagnosis are associated with disease-specific survival in parathyroid carcinoma. Annals of Surgical Oncology 25 25352540. (https://doi.org/10.1245/s10434-018-6559-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Medas F, Erdas E, Loi G, Podda F, Pisano G, Nicolosi A & Calò PG 2016 Controversies in the management of parathyroid carcinoma: a case series and review of the literature. International Journal of Surgery 28(Supplement 1) S94S98. (https://doi.org/10.1016/j.ijsu.2015.12.040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moher D, Shamseer L, Clarke M, Ghersi D, Liberati A, Petticrew M, Shekelle P, Stewart LA & PRISMA-P Group 2015 Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Systematic Reviews 4 1. (https://doi.org/10.1186/2046-4053-4-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mucci-Hennekinne S, Desolneux G, Luyckx F, Gibelin H, Mirallié E, de Calan L, Kraimps JL & Hamy A 2008 Carcinome parathyroïdien.: étude multicentrique de 17 patients. Journal de Chirurgie 145 133137. (https://doi.org/10.1016/s0021-7697(0873722-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Munson ND, Foote RL, Northcutt RC, Tiegs RD, Fitzpatrick LA, Grant CS, Heerden van JA, Thompson GB & Lloyd RV 2003 Parathyroid carcinoma: is there a role for adjuvant radiation therapy? Cancer 98 23782384. (https://doi.org/10.1002/cncr.11819)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nadarasa K, Bailey M, Chahal H, Raja O, Bhat R, Gayle C, Grossman AB & Druce MR 2014 The use of Cinacalcet in pregnancy to treat a complex case of parathyroid carcinoma. Endocrinology, Diabetes and Metabolism Case Reports 2014 140056. (https://doi.org/10.1530/EDM-14-0056)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • O’Neill CJ, Chan C, Symons J, Learoyd DL, Sidhu SB, Delbridge LW, Gill A & Sywak MS 2011 Parathyroid carcinoma encountered after minimally invasive focused parathyroidectomy may not require further radical surgery. World Journal of Surgery 35 147153. (https://doi.org/10.1007/s00268-010-0826-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pandya C, Uzilov AV, Bellizzi J, Lau CY, Moe AS, Strahl M, Hamou W, Newman LC, Fink MY & Antipin Y et al.2017 Genomic profiling reveals mutational landscape in parathyroid carcinomas. JCI Insight 2 e92061. (https://doi.org/10.1172/jci.insight.92061)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qian B, Qian Y, Hu L, Zhang S, Mei L & Qu X 2022 Prognostic analysis for patients with parathyroid carcinoma: a population-based study. Frontiers in Neuroscience 16 784599. (https://doi.org/10.3389/fnins.2022.784599)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Quinn CE, Healy J, Lebastchi AH, Brown TC, Stein JE, Prasad ML, Callender GG, Carling T & Udelsman R 2015 Modern experience with aggressive parathyroid tumors in a high-volume new England referral center. Journal of the American College of Surgeons 220 10541062. (https://doi.org/10.1016/j.jamcollsurg.2014.10.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ramos REO, Perez MM, Alves MFS, Piotto GHM, Takahashi TK, Gomes da Fonseca L, Silvino MCM, Hoff PM & de Castro G 2017 Malignancy-related hypercalcemia in advanced solid tumors: survival outcomes. Journal of Global Oncology 3 728733. (https://doi.org/10.1200/JGO.2016.006890)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Robert JH, Trombetti A, Garcia A, Pache JC, Herrmann F, Spiliopoulos A & Rizzoli R 2005 Primary hyperparathyroidism: can parathyroid carcinoma be anticipated on clinical and biochemical grounds? Report of nine cases and review of the literature. Annals of Surgical Oncology 12 526532. (https://doi.org/10.1245/ASO.2005.06.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Roukain A, Alwan H, Bongiovanni M, Sykiotis GP & Kopp PA 2021 Denosumab for the treatment of hypercalcemia in a patient with parathyroid carcinoma: a case report. Frontiers in Endocrinology (Lausanne) 12 794988. (https://doi.org/10.3389/fendo.2021.794988)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ruda JM, Hollenbeak CS & Stack BC Jr 2005 A systematic review of the diagnosis and treatment of primary hyperparathyroidism from 1995 to 2003. Otolaryngology–Head and Neck Surgery 132 359–372. (https://doi.org/10.1016/j.otohns.2004.10.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rubin MR, Bilezikian JP, Birken S & Silverberg SJ 2008 Human chorionic gonadotropin measurements in parathyroid carcinoma. European Journal of Endocrinology 159 469474. (https://doi.org/10.1530/EJE-08-0169)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ryhänen EM, Leijon H, Metso S, Eloranta E, Korsoff P, Ahtiainen P, Kekäläinen P, Tamminen M, Ristamäki R & Knutar O et al. 2017 A nationwide study on parathyroid carcinoma. Acta Oncologica 56 9911003. (https://doi.org/10.1080/0284186x.2017.1306103)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sadler C, Gow KW, Beierle EA, Doski JJ, Langer M, Nuchtern JG, Vasudevan SA & Goldfarb M 2014 Parathyroid carcinoma in more than 1,000 patients: a population-level analysis. Surgery 156 16221629. (https://doi.org/10.1016/j.surg.2014.08.069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sali AP, Motghare P, Bal M, Mittal N, Rane S, Kane S & & Patil A 2021 parathyroid carcinoma: a single-institution experience with an emphasis on histopathological features. Head and Neck Pathology 15 544554. (https://doi.org/10.1007/s12105-020-01244-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schulte KM & & Talat N 2012 Diagnosis and management of parathyroid cancer. Nature Reviews. Endocrinology 8 612622. (https://doi)

  • Schulte KM, Talat N, Miell J, Moniz C, Sinha P & Diaz-Cano S 2010 Lymph node involvement and surgical approach in parathyroid cancer. World Journal of Surgery 34 26112620. (https://doi.org/10.1007/s00268-010-0722-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schulte KM, Gill AJ, Barczynski M, Karakas E, Miyauchi A, Knoefel WT, Lombardi CP, Talat N, Diaz-Cano S & Grant CS 2012 Classification of parathyroid cancer. Annals of Surgical Oncology 19 26202628. (https://doi.org/10.1245/s10434-012-2306-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schulte KM, Talat N, Galata G, Gilbert J, Miell J, Hofbauer LC, Barthel A, Diaz-Cano S & Bornstein SR 2014 Oncologic resection achieving R0 margins improves disease-free survival in parathyroid cancer. Annals of Surgical Oncology 21 18911897. (https://doi.org/10.1245/s10434-014-3530-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schulte JJ, Pease G, Taxy JB, Hall C & Cipriani NA 2021 Distinguishing parathyromatosis, atypical parathyroid adenomas, and parathyroid carcinomas utilizing histologic and clinical features. Head and Neck Pathology 15 727736. (https://doi.org/10.1007/s12105-020-01281-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Selvan B, Paul MJ, Seshadri MS, Thomas N, Paul T, Abraham D, Oommen R, Shandhly N, John S & Rajaratnam S et al.2013 High index of clinical suspicion with optimal surgical techniques and adjuvant radiotherapy is critical to reduce locoregional disease progression in parathyroid carcinoma. American Journal of Clinical Oncology 36 6469. (https://doi.org/10.1097/COC.0b013e31823a4924)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shaha AR & Shah JP 1999 Parathyroid carcinoma: a diagnostic and therapeutic challenge. Cancer 86 378380. (https://doi.org/10.1002/(SICI)1097-0142(19990801)86:3<378::AID-CNCR3>3.0.CO;2-F)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shah R, Gosavi V, Mahajan A, Sonawane S, Hira P, Kurki V, Bal M, Sathe P, Pai P & D’Cruz A et al.2021 Preoperative prediction of parathyroid carcinoma in an Asian Indian cohort. Head & Neck 43 20692080. (https://doi.org/10.1002/hed.26677)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sidhu PS, Talat N, Patel P, Mulholland NJ & Schulte KM 2011 Ultrasound features of malignancy in the preoperative diagnosis of parathyroid cancer: a retrospective analysis of parathyroid tumours larger than 15 mm. European Radiology 21 1865–1873. (https://doi.org/10.1007/s00330-011-2141-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Silva-Figueroa A, Villalobos P, Williams MD, Bassett RL, Clarke CN, Lee JE, Busaidy NL & Perrier ND 2018 Characterizing parathyroid carcinomas and atypical neoplasms based on the expression of programmed death-ligand 1 expression and the presence of tumor-infiltrating lymphocytes and macrophages. Surgery 164 960964. (https://doi.org/10.1016/j.surg.2018.06.013)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Silverberg SJ, Rubin MR, Faiman C, Peacock M, Shoback DM, Smallridge RC, Schwanauer LE, Olson KA, Klassen P & Bilezikian JP 2007 Cinacalcet hydrochloride reduces the serum calcium concentration in inoperable parathyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 92 38033808. (https://doi.org/10.1210/jc.2007-0585)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stang A 2010 Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. European Journal of Epidemiology 25 603605. (https://doi.org/10.1007/s10654-010-9491-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Storvall S, Leijon H, Ryhänen EM, Vesterinen T, Heiskanen I, Schalin-Jäntti C & Arola J 2021 Filamin A and parafibromin expression in parathyroid carcinoma. European Journal of Endocrinology 185 803812. (https://doi.org/10.1530/EJE-21-0668)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Takeuchi Y, Takahashi S, Miura D, Katagiri M, Nakashima N, Ohishi H, Shimazaki R & Tominaga Y 2017 Cinacalcet hydrochloride relieves hypercalcemia in Japanese patients with parathyroid cancer and intractable primary hyperparathyroidism. Journal of Bone and Mineral Metabolism 35 616622. (https://doi.org/10.1007/s00774-016-0797-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Talat N & Schulte KM 2010 Clinical presentation, staging and long-term evolution of parathyroid cancer. Annals of Surgical Oncology 17 21562174. (https://doi.org/10.1245/s10434-010-1003-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tzotzas T, Goropoulos A, Karras S, Terzaki A, Siolos A, Doumas A, Zaramboukas T & Tigas S 2022 Effective long-term management of parathyromatosis-related refractory hypercalcemia with a combination of denosumab and Cinacalcet treatment. Hormones (Athens, Greece) 21 171176. (https://doi.org/10.1007/s42000-021-00343-w)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Uljanovs R, Strumfa I, Bahs G, Vidusa L, Merkurjeva K, Franckevica I & & Strumfs B 2021 Molecular profile of parathyroid tissues and tumours: a heterogeneous landscape. Polish Journal of Pathology 72 99116. (https://doi.org/10.5114/pjp.2021.109513)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vellanki P, Lange K, Elaraj D, Kopp PA & El Muayed M 2014 Denosumab for management of parathyroid carcinoma-mediated hypercalcemia. Journal of Clinical Endocrinology and Metabolism 99 387390. (https://doi.org/10.1210/jc.2013-3031)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Verdelli C & Corbetta S 2017 Epigenetic alterations in parathyroid cancers. International Journal of Molecular Sciences 18 310. (https://doi.org/10.3390/ijms18020310)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Villar-del-Moral J, Jiménez-García A, Salvador-Egea P, Martos-Martínez JM, Nuño-Vázquez-Garza JM, Serradilla-Martín M, Gómez-Palacios A, Moreno-Llorente P, Ortega-Serrano J & de la Quintana-Basarrate A 2014 Prognostic factors and staging systems in parathyroid cancer: a multicenter cohort study. Surgery 156 11321144. (https://doi.org/10.1016/j.surg.2014.05.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wächter S, Holzer K, Manoharan J, Brehm C, Mintziras I, Bartsch DK & & Maurer E 2019 Chirurgie des Nebenschilddrüsenkarzinoms. Der Chirurg 90 905912. (https://doi.org/10.1007/s00104-019-1007-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang O, Wang CY, Shi J, Nie M, Xia WB, Li M, Jiang Y, Guan H, Meng XW & Xing XP 2012 Expression of Ki-67, galectin-3, fragile histidine triad, and parafibromin in malignant and benign parathyroid tumors. Chinese Medical Journal 125 28952901.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang J, Wang Q, Zhao T, Liu X, Bai G, Xin Y, Shen H & & Wei B 2021 Expression profile of serum-related exosomal miRNAs from parathyroid tumor. Endocrine 72 239248. (https://doi.org/10.1007/s12020-020-02535-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang C, Wen K, Dai L, Wen S & & Zhang Y 2022 The clinical features and treatment strategy of parathyroid cancer: a retrospective analysis. BioMed Research International 23 1913900. (https://doi.org/10.1155/2022/1913900)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wei CH & Harari A 2012 Parathyroid carcinoma: update and guidelines for management. Current Treatment Options in Oncology 13 1123. (https://doi.org/10.1007/s11864-011-0171-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wei B, Zhao T, Shen H, Jin M, Zhou Q, Liu X, Wang J & & Qian Wang Q 2022 Extended en bloc reoperation for recurrent or persistent parathyroid carcinoma: analysis of 31 cases in a single institute experience. Annals of Surgical Oncology 29 12081215. (https://doi.org/10.1245/s10434-021-10962-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Witteveen JE, Hamdy NAT, Dekkers OM, Kievit J, van Wezel T, Teh BT, Romijn JA & Morreau H 2011 Downregulation of CASR expression and global loss of parafibromin staining are strong negative determinants of prognosis in parathyroid carcinoma. Modern Pathology 24 688697. (https://doi.org/10.1038/modpathol.2010.236)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xue S, Chen H, Lv C, Shen X, Ding J, Liu J & Chen X 2016 Preoperative diagnosis and prognosis in 40 parathyroid Carcinoma Patients. Clinical Endocrinology 85 2936. (https://doi.org/10.1111/cen.13055)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Young S, Wu JX, Li N, Yeh MW & Livhits MJ 2016 More extensive surgery may not improve survival over parathyroidectomy alone in parathyroid carcinoma. Annals of Surgical Oncology 23 28982904. (https://doi.org/10.1245/s10434-016-5256-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou L, Huang Y, Zeng W, Chen S, Zhou W, Wang M, Wei W, Zhang C, Huang J & Liu Z et al.2021 Surgical disparities of parathyroid carcinoma: long-term outcomes and deep excavation based on a Large database. Journal of Oncology 2021 8898926. (https://doi.org/10.1155/2021/8898926)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Adam MA, Untch BR & Olson JA 2010 Parathyroid carcinoma: current understanding and new insights into gene expression and intraoperative parathyroid hormone kinetics. Oncologist 15 6172. (https://doi.org/10.1634/theoncologist.2009-0185)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Agarwal S, Kardam S, Chatterjee P, Kumar C, Boruah M, Sharma MC, Tabin M & Ramakrishnan L 2022 CaSR expression in normal parathyroid and PHPT: new insights into pathogenesis from an autopsy-based study. Journal of Endocrinological Investigation 45 337346. (https://doi.org/10.1007/s40618-021-01646-w)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Al-Kurd A, Mekel M & Mazeh H 2014 Parathyroid carcinoma. Surgical Oncology 23 107114. (https://doi.org/10.1016/j.suronc.2014.03.005)

  • Apaydın T & & Yavuz DG 2021 Seven cases of parathyroid carcinoma and review of the literature. Hormones (Athens) 20 189195. (https://doi.org/10.1007/s42000-020-00220-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Asare EA, Sturgeon C,