Submit manuscript...
eISSN: 2473-0815

Endocrinology & Metabolism International Journal

Mini Review Volume 5 Issue 6

Parathyroid hormone related peptide (PTHrP): a mini-review

Michael AB Naafs1

1Dutch Internist Endocrinologist, Naafs International Health Consultancy, Netherlands
1Dutch Internist Endocrinologist, Naafs International Health Consultancy, Netherlands

Correspondence: Michael AB Naafs, Dutch Internist Endocrinologist, Health Consultant at Naafs International Health Consultancy, Rhodoslaan 20, 7577KN, Oldenzaal, The Netherlands, Tel +31681589079

Received: October 12, 2017 | Published: November 9, 2017

Citation: Naafs MAB. Parathyroid hormone related peptide (PTHrP): a mini-review. Endocrinol Metab Int J. 2017;5(6):321-328. DOI: 10.15406/emij.2017.05.00139

Download PDF

Abstract

In this mini-review the role of parathyroid hormone-related peptide (PTHrP) in physiology and pathophysiology is discussed. Evolving from an unknown humoral factor in the HHM (Hypercalcemia of malignancy) syndrome its role as a “masterregulator” in the PTH family following identification is reviewed. PTHrP has more paracrine and autocrine functions in virtually all tissues than classical endocrine functions and is essential in embryonic, fetal and post-natal life. The physiological functions are near endless. The development of PTH/PTHrP receptor antagonist and agonists has been difficult and disappointing. Its role as a possible cytokine itself and as a regulatory thermogenesis factor in brown adipose tissue in cancer cachexia is discussed. PTHrP assays must be improved for PTHrP to be a reliable biomarker in oncology and to resolve the function of local proteolytic PTHrP fragments which are largely a mystery until now.

Keywords: parathyroid hormone-related peptide, PTHrP, hypercalcemia, HHM, HHM syndrome

Abbreviations

PTHrP, parathyroid hormone-related peptide; PHP, primary hyperparathyroidism; MS, mass spectometry; HHM, hypercalcemia of malignancy

Introduction

In most clinical situations Hypercalcemia is caused by primary hyperparathyroidism (PHP) or malignancy.1,2 Because the excretion of urinary cAMP (cyclic 3’,5’ adenosine monophosphate) has been proved to be a good measurement of the biological activity of parathyroid hormone (PTH) in PHP some investigators started measuring urinary cAMP excretion in hypercalcemic patients with a malignant tumor in an attempt to differentiate between the Hypercalcemia of PHP and of malignancy.3-5 Soon it appeared that a considerable part of patients with Hypercalcemia of malignancy (HHM) also showed an increased urinary cAMP excretion. This was found in particular in hypercalcemic patients without bone metastases.6-8 Like in PHP these patients also had a decreased tubular reabsorption of phosphate (TmPO4/GFR). However in contrast to patients with PHP normal or low levels of 1.25. di-(OH)D3 were found and the tubular reabsorption of calcium was not increased.8

According to Stewart et al the humoral factor that leads to an increased urinary cAMP excretion and Hypercalcemia in patients with a malignancy without skeletal metastases is not identical to PTH and couldn’t be measured with a PTH radioimmunoassay. Nevertheless this humoral factor resembles PTH in decreasing TmPO4/GFR and increasing nephrogenous cAMP excretion. For that reason it was called a “PTH-like factor” resulting in Hypercalcemia in patients with malignancy and it was named the “Humoral Hypercalcemia of Malignancy (HHM) syndrome”.8 The HHM syndrome occured in particular in patients with squamous cell solid tumors of the bronchus, head and neck and in the renal adenocarcinoma.8

Ralston et al.9 showed there was no association between the number of skeletal metastases and the serum calcium concentration in patients with solid tumors. Based on this study Mundy estimated that the HHM syndrome occurs in 75% of patients with a solid tumor, including breast cancer patients.10 Hypercalcemia in malignancy is caused in 50% by breast cancer and multiple myeloma. Therefore it was important that the occurence of the HHM syndrome in breast cancer was demonstrated by Isales et al.11

Several investigators studied nephrogenous cAMP excretion in normocalcemic tumor patients and found nephrogenous cAMP excretion increased in 30% of patients, including breast cancer patients.6,12,13 If this is a harbinger for the development of Hypercalcemia is not known. What is well known that with resection of the tumor all biochemical parameters disappear. It is also known that the renal adenyl cyclase system in these patients shows a diminished stimulation capability.14-17 It was in the late 1980’s the “PTH-like factor” was identified as the Parathyroid Hormone Related Peptide (PTHrP).18-20 In this mini -review the identification, physiological and pathophysiological role and future possible deveopments of PTHrP will be discussed.

Identification of PTHrP

Human PTHrP proved to be encoded by a single gene on the short arm of chromosome 12.18-22 Alternative splicing generates many different mRNA species which encode three separate is forms of 139, 141 or 173 amino acids. From the outset it was evident that the PTHrP and PTH gene were related. The portions of the genes encoding the amino-terminal part are highly homologous such that the peptides share 8 of the first 13 amino acids and a similar secondary structure over the next 21 amino acids. The amino-terminal end of PTH contains 34 amino acids. PTHrP is produced in low concentrations in virtually all tissues. The physiological role of PTHrP remains incompletely understood. PTHrP has a fiunction in:

  1. Transepithelial calcium transport in the kidney and mammary gland.
  2. Smooth muscle relaxation in the uterus, bladder, gastrointestinal tract and arterial wall.
  3. Cellular differentiation and apoptosis of multiple tissues.
  4. PTHrP is an indispensible component of successful pregnancy and fetal development. Embryonic gene deletion is lethal in mammals.

When the receptor was discovered it was evident that this receptor functioned to relay both PTH and PTHrP biological activity.23,24 This PTH1 receptor belongs to the G protein-coupled receptor family. Knock out of this PTH/PTHrP receptor (PPR) showed the importance of PTHrP.25,26 PTHrP is the master regulator and has widespread paracrine actions. The acions of recombinant PTH 1-34, as an anabolic agent in the treatment of osteoporosis mimic PTHrPs actions locally in the bone microenvironment.27 There are only three identified circumstances in which PTHrP species are present in the circulation and act in an endocrine manner:

  1. The HHM syndrome in which PTHrP is produced by tumors and circulates to the bone to stimulate bone resorption.28,29
  2. Lactation in which PTHrP is made in the breast and reaches the circulation.30
  3. Fetal life, where PTHrP regulates maternal-to-fetal calcium transport.31,32

Hence the vast majorities of PTHrP actions are paracrine in nature.

Skeletal actions of PTHrP

The generation of mice missing the PTHrP gene, the PTH/PTHrP receptor gene or the PTH gene provided important models for testing the physiological roles of PTHrP.25,33,34 Mice lacking PTHrP die at birth because all bones resulting from endochondrial bone formation develop improperly. As a result the rib cage is small and inappropriately mineralized. Abnormalities in the growth plate result from abberant control of chondrocyte proliferation and differentiation in the PTHrP (-/-) growth plates and also in the growth plates of the PTH/PTHrP receptor (-/-) mices. These similarities support the idea that PTHrP affects the growth plate by activating the PTH/PTHrP receptor.35,36 This phenotype was not seen in mice with PTH deletion. Heterozygous mice with only one null PTHrP allele (PTHrP +/-) developed premature osteoporosis due to decreased bone formation linked to enhanced apoptosis of osteogenic cells.34,37 Exogenous application of PTH or PTHrP prevented apoptosis and associated bone loss.27

Recombinant PTH(1-34), teriparatide (Forteo) is approved for the treatment of osteoporosis but may cause bone resorption, hypercalcemia, nausea, muscle cramps and other adverse effects.38 In contrast a study with the 1-36 amino- terminal peptide of PTHrP didn’t activate bone resorption and acted as a pure anabolic agent while causing none of the adverse effects of teriparatide. Although intermittent treatment with PTHrP causes an anabolic response continous treatment causes paradoxical downregulation of osteogenic genes resulting in skeletal catabolism.39,40 PTH(1-34) and PTHrP(1-36) bind the PTH1 receptor with approximately equal affinity while shorter amino terminal fragments do not. This activity reflects the strong similarity of the secondary and tertiary structure despite the differences in primary amino acid sequence in this region.41

Other mouse genetic studies were also helpful in discerning differences in PTH and PTHrP physiology. Global knockout of PTH in the mouse resulted in hypocalcemia and hyperphosphatemia and a bone phenotype of increased trabecular and cortical bone volume the opposite of PTHrP null mice of the same age.34 This suggests that PTH physiologically does not function to promote bone formation but rather acts as a regulator of extracellular fluid calcium postnatally and in the fetus and is essential for providing calcium for mineralization of bone. The fact that PTH null mice remained hypocalcemic and when crossed with PTHrP(+/-) mice shows the lack of a calcium regulatory role for PTHrP. Its physiological role after development is that of a local regulator of bone remodeling, formation and resorption.

Mice lacking both the midregion NLS (nuclear localizing site) and the carboxy terminal part after knock-in PTHrP(1-84) while retaining the bioactive amino terminal region showed multiple abnormalities and early lethality at 2-3 weeks age. Homozygous mice showed skeletal retardation and decreased bone mass with reduced proliferation of osteoblasts and increased apoptosis of osteoblasts. Together with fewer osteoblasts there was less trabecular and cortical bone and fewer osteoclasts. Impaired growth plate chondrocyte proliferation resulted in mice with markedly shorter long bones. They differed quite clearly from the severe chondrodystrophy of PTHrP null mice, which resulted from premature differentiation of chondrocytes.35 The PTHrP (1-84) knock-in mice exhibited also early senescence in multiple tissues. The brain showed decreased neural proliferation and increased apoptosis with abnormal shape. PTHrP is needed in maintaining normal synapsis and plasticity. These effects are partly mediated by the PTH2 receptor.42

In a second model, knock-in of PTHrP(1-66), which excludes a significant portion of the mid-region, resulted in a an even more severe phenotype with similar skeletal abnormalities as chondrodysplasia but also impaired hemopoiesis and mammary development, dysregulated energy metabolism and death by 5 days of age. So many actions of PTHrP are not mediated solely by the amino-terminal fragment.43

PTHrP in lactation

Not long after its discovery PTHrP mRNA was found in the lactating and PTHrP was measured in high concentrations in milk44,45 In mice as soon as the mammary bud begins to form epithelial cells produce PTHrP which interacts with the PTH1 receptor expressed on surrounding mesenchymal cells. This interaction is necessary for proper differentiation of the dense mammary mesenchym. PTHrP or PTH1 R knock-out mice lack mammary glands. The formation of the human breast in human fetesus is similar to that in mice and PTHrP is indispensible for that as well.46 During puberty PTHrP seems to regulate the growth of the mammary ducts in response to estrogen.47,48 PTHrP is also made by breast epithelial cells during lactation and large quantities are secreted in milk.49,50 Milk production requires a great deal of calcium, an important source of which is the maternal skeleton. During lactation elevated levels of PTHrP correlate with bone loss in human and circulating levels of PTHrP correlate directly with rates of bone resorption and inversely with bone mass in mice.51,52 The lactating breast also expresses the CaR which signals to suppress PTHrP secretion in response to calcium delivery to the breast.52 These interactions define a classical negative feedback loop whereby mammary cells secrete PTHrP to mobilize calcium from the bone. Calcium in turn feeds back to inhibit further PTHrP secretion from the breast. Therefore during lactation the breast and bone engage in a conversation which leads to the mobilization of skeletal calcium to ensure a steady supply of calcium for milk production. Interestingly fish PTHrP shows a similar function to mobilize calcium stored in scales to be used for egg production.53

PTHrP and the placenta

During pregnancy calcium must be actively transported across the placenta from mother to fetus. The calcium concentration in the fetus is higher than in the mother so that calcium must be transported against a gradient.54 In PTHrP(-/-) mice this gradient is lost and PTHrP deficient fetuses are hypocalcemic. This suggests that fetal PTHrP is important in placental calcium transport from the mother.32 Placental production of PTHrP has been shown to be regulated by the CaR.55,56 Experiments in sheep and mice showed that the midregion PTHrP is responsible for placental calcium transport and not the amino-terminal portion.32,57 PTHrP levels in umbillical cord blood are approximately 10 fold higher than PTH levels. Placental calcium transfer requires PTHrP and possibly PTH but not calcitriol or calcitonin. It is in the neonatal period that intestinal calcium absorption and thereby skeletal development and mineralization become dependent upon vitamin D3 and calcitriol.58 A few cases of overproduction of PTHrP in the placenta resulting in hypercalcemia have been described.59,60

PTHrP in smooth muscle and the cardiovascular system

PTHrP expression is present in many different smooth muscle cell beds.22 Stretching of the muscular cell or structure increases the expression of PTHrP. PTHrP works as a relaxant then in turn on the structure that has been stretched in an autocrine or paracrine way.22,61,64 For organs like the bladder, stomach or uterus this may be an important feedback loop allowing gradual filling. In the vascular wall PTHrP is expressed by vasoconstrictive agents, stretch it and acts as a vasodilator to resistance vessels. In this way PTHrP may act as a local modulator of blood flow.65 PTHrP expression is also stimulated by pathologic stimuli including atherosclerosis, restenosis after balloon angioplasty and hypertension.66 In addition to its relaxant effect PTHrP inhibits vascular smooth muscle cell proliferation through its interaction with the PTH1R and stimulation of the cAMP-protein kinase a pathway resulting in cell cycle blockade in the G1 phase. These effects are mediated by the amino-terminal fragments of PTHrP. PTHrP may also have proliferative effects on vascular smooth muscle cells by translocating in the nucleus leading to inhibition of vascular proliferation. These effects are mediated by the mid-region fragments of PTHrP.67,68 Endogenous PTHrP is mitogenic in the renovascular smooth muscle cell while exogenous added PTHrP was anti-mitogenic in the SHR (Spontanously Hypertensive Rat) model of genetic hypertension. These results suggest that a single molecule may have opposite effects under physiological and pathological conditions.69 PTHrP has been found in cardiomyocytes and co-localizes with atrial natriuretic peptide in granules within the atrial cells in the rat heart.70 In PTH1R deficient mice widespread cardiomyocyte death occurred in midgestation. This was not seen in PTHrP(-/-) mice. PTHrP has both positive inotropic and chronotropic effects and may affect coronary blood flow.71,72

PTHrP and the pancreas

PTHrP is expressed in all four different types of neuroendocrine cells in the pancreas. In beta-cells it is stored within secreting granules and co-released with insulin.73 Pancreatic islet cells express the PTH1 receptor. Overexpression of PTHrP in beta cells leads to an increased beta cell mass, hyperinsulinemia and hypoglycemia due to increased beta cell proliferation, increased insulin production and inhibition of beta cell apoptosis.74,75 PTHrP(-/-) mice have no defects in islet cell development. Because of the death of these mice at birth the role of PTHrP in the physiology of islets is not known. Neuroendocrine tumors secreting PTHrP resulting in hypercalcemia has been described.76,77,78

PTHrP and the central and peripheral nervous system

PTHrP and the PTH1 receptor are both widely expressed within the brain including the cortex, the cerebellum, the hippocampus, hypothalamus and pituitary.79,80 PTHrP secretion by neurons is regulated by calcium influx through L-type channel activity on depolarization. In turn PTHrP can dampen L-type channel activity to protect neurons from damage to prolonged or repeated depolarization so called “excitotoxicity” creating an autocrine or paracrine short feedback loop.81 PTHrP is also present in glia and astrocytes. High PTHrP levels in glial tumors correlate with a poor prognosis.82-85 PTHrP expression increases in sites of ischemic brain injury where it may play a protective role by enhancing blood flow.83 PTHrP inhibits the proliferation of damaged and dedifferentiated Schwann cells in peripheral nerves in this way contributing to nerve regeneration.86 The brain contains also PTH2 receptors. These receptor scan bind PTH and the tuberoinfindibilar peptide (TIP39) but do not bind PTHrP. The TIP39-PTH2R neuromodulatory system is predominantly involved in auditory processing and in neuroendocrine modulation. TIP39 influences somatostatin and CRH (Corticotrophin Releasing Hormone) neurons. PTHrP has no role in this system.87

Teeth and PTHrP

Developing teeth are surrounded by bone and must erupt through the roof of the dental crypt to emerge into the oral cavity. This requires spatial coordination of bone cell activity. Osteoclasts must resorb the bone overlying the crown of the tooth to allow it to emerge and osteoblasts must form bone at the base of the tooth to propel it upward out of the crypt. PTHrP is produced by stellate reticulum cells and it signals to dental follicle cells to promote the formation of osteoclasts above the crypt. In the absence of PTHrP these osteoclasts do not appear, eruption fails to occur and the teeth become impacted.88,90

PTHrP in the HHM sydrome

After the identification of PTHrP an assay had to be developed. PTHrP is cleaved into N-terminal (1-86) and C terminal.109-141 peptides. Various attempts have been made to develop assays that target the N-terminal or C-terminal peptide. The original sandwich radioimmunoassay was using two different antibodies targeting differerent PTHrP peptides showed to be specific for PTHrP but has now been discontinued (Nichols Lab). Quest offers currently a C-terminal PTHrP assay. This assay is however impacted by declining renal function producing higher values and more false positive results. N-terminal fragments are less influenced by renal function, producing less false positive results. For this reason detection of the N-terminal is a more useful indicator of HHM. Mayo Medical Laboratories developed a sandwich assay capturing the mid-region and the N-terminal fragment of PTHrP. Beckman offers a similar assay that captures the PTHrP 1-86 sequence. Others developed liquid-chromatography mass spectometry (MS) assays for the midportion region of PTHrP but it had very poor correlation with the immunoassay. Competetive immunoassays have lower specificity, precision and analytical sensitivity compared to sandwich or MS assays. So when PTHrP should be a reliable biomarker there is still some work to do. At this time nephrogenous cAMP excretion remains the golden standard as a bio-assay until the PTHrP or other protein assays are able to achieve maximal clinical sensitivity and specificity.91

PTHrP/PTH1R antogonists and agonists

The development of PTHrP or PTHrP/PTH1R antagonists and agonists has been difficult and disappointing. Binding modes used by agonists and antagonists of the PTH1 receptor utilizes a two-site binding site mechanism that involves the putatively helical N-and C-terminals of the ligand. The N-terminal domain of the receptor provides the major binding energy while the juxtamembrane region of the receptor induces activation of the receptor. N-terminally truncated antagonists as PTHrP(5-36) bind mainly to the N-domain with some overlap to the J-domain. These N-terminal antagonists are not effective inhibitors of the two-site agonists such as PTH(1-34). Recently however highly potent and longacting PTH1R monoclonal antibodies were developed for the treatment of elevated levels of PTH as in primary hyperparathyroisism (PHP) or PTHrP as in the HHM syndromePTH1R antagonism to both PTH and PTHrP was determined by cAMP accumulation in osteosarcoma cell lines of both human and rat origin. This antibody showed roughly equivalent inhibition of cAMP in human and murine cell Lines. The antibody inhibited both PTH and PTHrP induced osteoclast differentiation by tenfold. The expression of RANKL was also inhibited. The monoclonal antibody reduced serum calcium levels in a dose dependent manner when infused.92-94 These studies were scheduled in the poster sessions of the Endocrine Society and the American Society of Cancer Research and not in the oral. Presentations implying clinical application of this monoclonal antibody will take some time.

As depicted above the physiologically functions of PTHrP are enomous and widespread. Systemic application of an anti-PTH/PTHrP receptor antibody could have also widespread adverse effects for that reason. Topical administration of such an antogonist would be less risky.PTH and PTHrP influence hair follicles through paracrine and intracranial routes. The PTH/PTHrP receptor plays an important role in the hair follicle cycle and may induce premature catagen-telogen transition.95 Transgenic mice with an overexpression or blockade (PTH/PTHrP receptor knock-out mice) of PTHrP activity revealed lessened or increased hairgrowth respectively. Antagonists of the PTH/PTHrP receptor have been shown to stimulate proliferation of hair follicle cells and hair growth.96 A hair stimulating effect of such an antagonist has been observed in hairless mice and mice treated with cyclofosfamide.96-98 So these topical PTH/PTH1R antagonists might be useful in chemotherapy induced alopecia.98 Sporadic cases of PTH-related hair symptoms were reported in familial syndromes of hyper/hypoparathyroidism.99,100 PTHrP antagonists have been studied also in enchondroma’s, patients with Olliers syndrome, chondroblastoma’s and chondrosarcoma’s. Research with structurally modified PTH1R ligands has revealed deeper insights in the function of the receptor but didn’t result in clinical applications yet.102

The role of PTHrP in the “cytokine excess” has been investigated too.As PTH and PTHrP bind to the PTH1R and stimulate osteoblasts via A-kinase IL-6(interlekine-6) and TNF alpa (tumor necrosis factor alpha) production is also stimulated by this pathway. Marked elevated levels of IL-6 and TNF alpha are present in PHP and in the HHM syndrome. These increases in cytokines normalize after parathyroidectomy.103 Levels of circulating cytokines as IL-6. IL-8 and TNF-alpha are also increased in cancer patients with high PTHrP levels.104 PTHrP has been found to be a key factor not only in HHM but also in local osteolytic Hypercalcemia in myeloma patients and osteolytic bone metastases.105,106 If PTHrP is a cytokine itself or only stimulates the production of the interleukins and TNF alpha is not known. How they interplay is also not completely understood. For PTHrP as well as the cytokines the RANKL-RANK pathway represents a final common pathway to osteoclastogenesis and stimulation of bone resorption.107 Bone remodelling is a continous process achieved by osteoblasts and osteoclasts. The balance of this process is maintained by a trimolecular control factor complex composed of osteoprotegerin (OPG), RANKL (osteoprotegerin ligand) and RANK (receptor activator of nuclear kappa beta, NFKB ligand). Osteocytes regulate the recruitment of osteoclasts to sites of bone resorption by inducing the expression of RANKL by osteoblastic cells. This system is part of the TNF family and can be unregulated by PTHrP, TNF alpha or macrophage colony stimulating factor (M-CSF).108,109

Denosumab is a fully human IG -G2 monoclonal neutralizing antibody to RANKL. It differs from bisphosphonates that inhibit bone resorption by inducing apoptosis in mature osteoclasts. Denosumab inhibits not only bone resorption by mature osteoclasts but also osteoclast formation from hematopoetic precursors.110-112 Denosumab was effective in controlling serum calcium levels in patients with HHM and was approved by the FDA for the treatment of skeletal metastases in malignancy, HHM and postmenopausal osteoporosis based on the trials delivered.113,114

PTHrP in cancer cachexia
In 2014 the Spiegelman group suggested that PTHrP is a mediator of adipose tissue browning and tumor-induced cachexia.115 PTHrP drives the expression of thermogenesis in adipose tissue. Neutralizing of PTHrP in tumor-bearing mice blocked adipose tissue browning and the loss of muscle mass and strenght. Cachexia is a wasting syndrome that accompanies many chronic diseases. The PTH/PTHrP receptor was shown also mediating the cachexia of chronic kidney failure. PTHrP proved to be involved in stimulating athermogenic gene program in 5/6 nephrectomized mice that suffered from cachexia. Fat-specific knock-out of PTH1R blocked adipose tissue browning and wasting. Loss of PTH1R in fat tissue preserved muscle mass and improved muscle strength. So PTHrP and PTH mediate wasting through a common mechanism involving a crosstalk between wasting of fat tissue and skeletal muscle.116,117 Patients with upper gastrointestinal and pancreatic cancers are the likeliest to develop cachexia which affects about 80% of terminal cancer patients.118 Blocking the “excess cytokines” syndrome could not reverse cachexia in terminal cancer patients as did the use of anabolic steroids neither.119 Blocking the PTH/PTHrP receptor in cancer cachexia will not be the ultimate answer because of the widespread systemic adverse effects to be expected as discussed before.

Conclusion

Evolving from an unknown humoral factor in the ”hypercalcemia of malignancy” (HHM) syndrome in the late 1970’s and 1980’s to the identification of this factor as “parathyroid hormone related peptide” (PTHrP) in 1987 has been a major step in the research of the function of members of the PTH-family in physiology and pathophysiology. Soon it appeared that most functions of PTHrP are paracrine or autocrine. Only its role in the HHM syndrome, lactation and in placental maternal-fetal calcium transport is classical endocrine in nature. PTHrP regulates endochondrial bone formation in fetal life and bone remodeling, formation and resorption in post-natal life. During lactation PTHrP provides a steady supply of calcium from the maternal skeleton to assure calcium-rich milk production. PTHrP is essential in pregnancy for regulating fetal-maternal calcium transport. Without PTHrP there is no fetal life. In the cardiovascular system PTHrP works as a vascular relaxant by its effect on the smooth muscles in the vessel wall and has it a chronotropic and inotropic effect on the heart .In the pancreas PTHrP is involved in beta cell proliferation, insulin production and inhibition of beta cell apoptosis. In the central nervous system PTHrP has a role in protecting neurons from damage of “excititoxicity” due to repeated or prolonged depolarization. Without PTHrP teeth will not erupt and become impacted in early life. PTHrP has also a function in the growth of the hair follicle. Antagonsts of the PTH/PTHrP receptor stimulate the proliferation of hair follicle cells and may be used in the future for chemotherapy-induced alopecia. PTHrP plays a role in the “cytokines excess” syndrome in cancer patients and may be a cytokine in itself. By bringing a thermogenesis gene program to expression in brown adipose tissue PTHrP plays a role in cancer cachexia too. Blocking excess cytokines does not relieve cancer cachexia and blocking the PTH/PtHrP receptor wills not either because of the expected widespread adverse effects. When PTHrP should be a reliable tumor marker in oncology problems with the specificity, sensitivity and analytical precision of the PTHrP assays must be resolved in the near future. This is rather urgent as the function of PTHrP fragments formed by local proteolysis stays otherwise largely unknown.

Conflicts of interest

None.

References

  1. Fisken RA, Heath DA, Bold AM. Hypocalcaemia‒a hospital review. Quart J Med. 1980;49(196):405‒418.
  2. Fisken RA, Heath DA, Somers S, Bold AM. Hypocalcaemia in hospital patients:clinical and diagnostic aspects. Lancet. 1981;1(8213):202‒207.
  3. Dohan PH, Yamashita K, Larsen P, et al. Evaluation of urinary cyclic 3’,5’‒adenosine monophosphate excretion in the differential diagnosis of hypercalcaemia. J Clin Endocrinol Metab. 1972;35(6):775‒784.
  4. Drezner MK, Neelan FA, Curtis HB, et al. Renal cyclic adenosine monophosphate: an accurate index of parathyroid function. Metabolism. 1976;25(10):1103‒1112.
  5. Murad F, Pak ChYC. Urinary excretion of adenosine 3’,5’‒monophosphate and guanosine 3’,5’‒monophosphate. N Engl J Med. 1972;286(26):1383‒1387.
  6. Rude RK, Sharp CF, Fredericks RS, et al. Urinary and nephrogenous adenosine 3’, 5’‒monophosphate in the hypocalcaemia of malignancy. J Clin Endocrinol Metab. 1981;52(4):765‒771.
  7. Rude RK, Sharp CF, Oldham SB. et al. Plasma cAMP, urinary cAMP and nephrogenous cAMP in the hypocalcaemia of malignancy. Clin Res. 1978;26:427.
  8. Stewart AF, Horst R, Deftos LJ, et al. Biochemical evaluation of patients with cancer associated with hypocalcaemia: evidence for humoral and non‒humoral groups. N Engl J Med. 1980;303(24):1377‒1383.
  9. Ralston SH, Gardner MD, Fogelman I, et al. Hypocalcaemia and metastatic bone disease: is there a causal link? Lancet. 1982;2(8304):903‒905.
  10. Mundy GR. Pathogenesis of hypocalcaemia of malignancy. Clin Endocrinol. 1985;23(6):705‒714.
  11. Isales C, Carcangiu ML, Stewart AF. Hypocalcaemia in breast cancer: assessment of the mechanism. Am J Med. 1987;82(6):1143‒1147.
  12. Abramson EC, Kukreja SC, Lad TE, et al. Nephrogenous cyclic adenosine monophosphate levels in normocalcaemic cancer patients: its significance. Horm Metab Res. 1984;16(4):204‒207.
  13. Van der Velden PC, Naafs MA, Fischer HR, et al. The urinary excretion of cAMP in patients with malignant tumors. Neth J Med. 1983;26(1):10‒17.
  14. Goltzman D, Stewart AF, Broadus AE. Malignancy‒associated hypocalcaemia: evaluation with a cyto‒chemical bioassay for parathyroid hormone. J Clin Endocrinol Metab. 1981;53(5):899‒904.
  15. Seshadri MS, Mason RS, Cornish CJ, et al. Humoral hypocalcaemia of malignancy: report of two further patients with biochemical studies on tumor extracts. Clin Sci (Lond). 1986;71(3):261‒269.
  16. Ralston SH, Boyce BF, Cowan RA, et al. Humoral hypocalcaemia of malignancy: metabolic and histo morphometric studies during surgical removal of the primary tumor. Q J Med. 1986;58(277):325‒335.
  17. Naafs MA, Fischer HR, van der Velden PC, et al. End‒organ resistance to PTH‒infusion in hypercalcaemic and normocalcaemic patients with solid tumours. Acta Endocrinol (Copenh). 1986;113(4):543‒550.
  18. Suva LJ, Winslow GA, Wettenhall RE, et al. A parathyroid hormone‒related protein implicated in malignant hypercalcemia: cloning and expression. Science. 1987;237(4817):893‒896.
  19. Burtis WJ, Wu T, Bunch C, et al. Identification of a novel 17.000 Dalton parathyroid hormone‒like related adenylate cyclase‒stimulating protein from a tumor associated with humoral hypocalcaemia of malignancy. J Biol Chem. 1987;262(15):7151‒7156.
  20. Strewler GJ, Stern PH, Jacobs JW, et al. Parathyroid hormone‒like protein from human renal carcinoma cells:Structural and functional homology with parathyroid hormone. J Clin Invest. 1987;80(6):1803‒1807.
  21. Mangin M, Webb AC, Dreyer BE, et al. An identification of a cDNA encoding a parathyroid hormone‒like peptide from a human tumor associated with humoral hypocalcaemia of malignancy. Proc Natl Acad Sci USA. 1988;85(2):597‒601.
  22. Philbrick WM, Wysolmerski JJ, Galbraith S, et al. Defining the roles of parathyroid hormone‒related protein in normal physiology. Physiol Rev. 1966;76(1):172‒173.
  23. Brewer HB, Fairwell T, Ronan R, et al. Human Parathyroid Hormone: Amino Acid Sequence of the Amino‒Terminal Residues 1‒34. Proc Natl Acad Sci USA. 1972;69(12):3585‒3588.
  24. Jüppner H, Abou‒Samra AB, Freeman M, et al. A G protein‒linked receptor for parathyroid hormone and parathyroid hormone related peptide. Science. 1991;254(5034):1024‒1026.
  25. Lanske B, Karaplis AC, Lee K, et al. PTH/PTHrP receptor in early development and Indian hedgehog regulated bone growth. Science. 1996;273(5275):663‒666.
  26. Lanske B, Divieti P, Kovacs CS, et al. The parathyroid hormone PTH/PTH‒related peptide receptor mediates actions of both ligands in murine bone. Endocrinology. 1998;139(12):5194‒5204.
  27. Miao D, He B, Jiang Y, et al. Osteoblast‒derived PTHrP is a potent endogenous bone anabolic agent that modifies the therapeutic efficacy of administered PTH 1‒34. J Clin Invest. 2005;115(9):2402‒2411.
  28. Burtis WJ, Fodero JP, Gaich G, et al. Preliminary characterization of circulating amino‒and carboxyl‒terminal fragments of parathyroid hormone related peptide in humoral hypocalcaemia of malignancy. J Clin Endocrinol Metab. 1992;75(4):1110‒1114.
  29. Grill V, Ho P, Body JJ, et al. Parathyroid hormone‒related protein: elevated levels in both humoral hypocalcaemia of malignancy and hypocalcaemia complicating metastatic breast cancer. J Clin Endocrinol Metab. 1991;73(6):1309‒1315.
  30. Grill V, Hillary J, Ho PM, et al. Parathyroid hormone‒related protein: a possible endocrine function in lactation. Clin Endocrinol. 1992;37(5):405‒410.
  31. Rodda CP, Kubota M, Heath JA, et al. Evidence for a novel parathyroid hormone‒related protein in fetal lamb parathyroid glands and sheep placenta: comparisons with a similar protein implicated in humoral hypercalcaemia of malignancy. J Endocrinol. 1988;117(2):261‒271.
  32. Kovacs CS, Lanske B, Hunzelman JL, et al. Parathyroid hormone‒related peptide (PTHrP) regulates fetal‒placental calcium transport through a receptor distinct from the PTH/PTHrP receptor. Proc Natl Acad Sci USA. 1996;93(26):15233‒15238.
  33. Karaplis AC, Luz A, Glowacki J, et al. Lethal skeletal dysplasia from targeted disruption of the parathyroid hormone‒related peptide gene. Genes Dev. 1994;8(3):277‒289.
  34. Miao D, He B, Karaplis AC, et al. Parathyroid hormone is essential for normal bone formation. J Clin Invest. 2002;109(9):1173‒1182.
  35. Amizuka N, Warshawsky H, Henderson JE, et al. Parathyroid hormone‒related peptide depleted mice show abnormal epiphyseal cartilage development and altered endochondral bone formation. J Cell Biol. 1994;126(6):1611‒1623.
  36. Lee K, Lanske B, Karaplis AC, et al. Parathyroid hormone‒related peptide delays terminal differentiation of chondrocytes during enchondral bone development. Endocrinology. 1996;137(11):5109‒5118.
  37. Amizuka N, Karaplis AC, Henderson JE, et al. Haplo insufficiency of parathyroid hormone‒related peptide (PTHrP) results in abnormal postnatal bone development. Develop Biol. 1996;175(1):166‒176.
  38. Horwitz MJ, Tedesco MB, Garcia Ocaña A, et al. Parathyroid Hormone related Protein for the Treatment of Postmenopausal Osteoporosis: Defining the Maximal Tolerable Dose. J Clin Endocrinol Metab. 2010;96(3):1279‒1287.
  39. Hardwick T, Kalia P, Baawa Ameyew J. Continued vs Intermittent Cell Exposure to Parathyroid Hormone‒Related Peptide on Osteogenic and Angiogenic Gene Expression in Human Osteoblasts and Bone Marrow Stromal Cells. Bone Joint J Orthoped Proceedings. 2013;95(Suppl 13):24.
  40. De Cortazar A, Alonso V, Alvarez Arogo M. Transient exposure to PTHrP(107‒139) exerts anabolic effects through vascular endothelial growth factor receptor 2 in human osteoblastic cells in vitro. Calc Tissue Int. 2006;79(5):360‒369.
  41. Orleff J, Wu TL, Stewart AF. Parathyroid hormone‒like proteins: biochemical responses and receptor interactions. Endocr Rev. 1989;10(4):476‒495.
  42. Gu Z1, Liu Y, Zhang Y, et al. Absence of PTHrP nuclear localization and carboxy terminus sequences leads to abnormal brain development and function. Plos One. 2012;7(7):e1542.
  43. Toribio RE, Brown HA, Novince CM, et al. The midregion, nuclear localization seguence and C terminal of PTHrP regulate skeletal development, hemopoiesis and survival in mice. FASEB J. 2010;24(6):1947‒1957.
  44. Budayr AA, Halloran BR, Halloran, et al. High levels of a parathyroid hormone‒like protein in milk. Proc Natl Acad Sci. 1989;86(7):7183‒7185.
  45. Thiede MA, Rodan GA. Expression of a calcium‒mobilizing parathyroid hormone‒like peptide in lactating mammary tissue. Science. 1988;242(4876):278‒280.
  46. Wysolmerski JJ, Cormier S, Philbrick WM, et al. Absence of functional type 1 parathyroid hormone (PTH) PTH related protein receptors in humans is associated with abnormal breast development and tooth impaction. J Clin Endocrinol Metab. 2001;86:1788‒1794.
  47. Dunbar ME, Dann P, Brown CW, et al. Temporally regulated over expression of parathyroid hormone‒related protein in the mammary gland reveals distinct fetal and pubertal phenotypes. J Endocrinol. 2001;171:403‒416.
  48. Dunbar ME, Young P, Zhang JP, et al. Stromal cells are critical targets in the regulation of mammary ductal morphogenesis by parathyroid hormone‒related protein (PTHrP). Dev Biol. 1998;203(1):75‒89.
  49. VanHouten JN, Dann P, Stewart AF, et al. Mammary specific deletion of parathyroid hormone‒related protein preserves bone mass during lactation. J Clin Invest. 2003;112(9):1429‒1436.
  50. Budayr AA, Halloran BR, Halloran, et al. High levels of a parathyroid hormone‒like protein in milk. Proc Natl Acad Sci USA. 1989;6:7183‒7185.
  51. Sowers MF, Hollis BW, Shapiro B, et al. Elevated parathyroid hormone‒related peptide associated with lactation and bone loss. JAMA. 1996;276(6):549‒554.
  52. Van Houten JN, Wysolmerski JJ. Low estrogen and high parathyroid hormone‒related peptide levels contribute to accelerated bone resorption and bone loss in lactating mice. Endocrinology. 2003;144(12):5521‒5529.
  53. Guirreiro PM, Renfro JL, Power DM. The parathyroid hormone family of peptides. Structure, tissue distribution, regulation and potential functional roles in calcium and phosphate balance in fish. Am J Physiol Regul Integr Comp Physiol. 2007;292(2):679R‒696R.
  54. Kovacs CS. Calcium and bone metabolism in pregnancy and lactation. J Clin Endocrinol Metab. 2001;86(6):2344‒2348.
  55. Hellman P, Ridefelt P, Juhlin C, et al. Parathyroid‒like regulation of parathyroid hormone‒related protein release and cytoplasmic calcium in cytothrophoblast cells of human placenta. Arch Biochem Biophys. 1992;293(1):174‒180.
  56. Kovacs CS, Ho C, Seidman CE. Parathyroid calcium sensing receptor regulates fetal blood calcium and fetal‒maternal calcium gradient independently of the maternal calcium levels. J Bone Min Res. 1996;22:S121.
  57. Care AD, Abbas SK, Pickard DW, et al. Stimulation of ovine placental transport of calcium and magnesium by mid‒molecule fragments of human parathyroid hormone‒related proteins. Exp Physiol. 1990;75(4):605‒608.
  58.  Kovacs CS. Bone metabolism in the fetus and neonate. Pediatr. Nephrol. 2014;29(5):793‒803.
  59. Sato K. Hypercalcemia during pregnancy and lactation and a case report of hypercalcemic crisis after delivery due to excessive production of PTH‒related protein (PTHrP) without malignancy (humoral hypercalcemia of pregnancy). Endocr J. 2008;55(6):959‒966.
  60. Eller Vainicher C, Ossola MW, Beck Peccoz P, et al. PTHrP‒associated hypercalcemia of pregnancy resolved after delivery: a case report. Eur J Endocrinol. 2012;166(4):753‒756.
  61. Strewler GJ. The physiology of parathyroid hormone‒related protein. N Engl J Med. 2000;342(3):177‒185.
  62. Thiede MA, Daifotis AG, Weir AC. Intrauterine occupancy controls expression of the parathyroid hormone‒related peptide gene in preterm rat myometrium. Proc Natl Acad Sc USA. 1990;87:6669‒6673.
  63. Thiede MA, Harm SC, McKee RL, et al. Expression of the parathyroid hormone‒related protein gene in the avian oviduct: Potential role as a local modulator of vascular smooth muscle tension and Shell gland motility during the egg‒ laying cycle. Endocrinol. 1991;129(4):1958‒1966.
  64. Yamamoto M, Harm SC, Grasser WA, et al. Parathyroid hormone‒related protein in the rat urinary bladder: A smooth muscle cell relaxant produced locally in response to mechanical stretch. Proc Natl Acad Sci USA. 1992;89(12):5326‒5330.
  65. Massfelder T, Helwig JJ. Parathyroid hormone‒related protein in cardiovascular development and blood pressure regulation. Endocrinology. 1999;140(4):1507‒1510.
  66. Massfelder T, Helwig JJ. The parathyroid hormone‒related protein system: More data but more unresolved questions. Curr Opin Nephrol Hypert. 2003;12(1):35‒42.
  67. Stuart WD, Maeda S, Khera P, et al. Parathyroid hormone‒related protein induces G1 phase arrest of vascular smooth muscle cells. Am J Physiol Endocrinol Metab. 2000;297(1):60‒67.
  68. Massfelder T, Dann P, Wu TL, et al. Opposing mitogenic and antimitogenic actions of parathyroid‒hormone‒related protein in vascular smooth muscle cells. A critical role for nuclear targeting. Proc Natl Acad Sci USA. 1997;94(25):13630‒13635.
  69.  Thierry M, Nathalie T, Nicole E, et al. Paradoxical actions of exogenous and endogenous parathyroid hormone‒related protein on renal vascular smooth muscle cell proliferation: reversion in the SHR model of genetic hypertension. FASEB J. 2001;15(3):707‒718.
  70. Deftos LJ, Burton DW, Brandt DW. Parathyroid hormone‒like protein is a secretory product of atrial myocytes. J Clin Invest. 1993;92(2):727‒735.
  71. Qian J, Colbert MC, Witte D, et al. Midgestational lethality in mice lacking the parathyroid hormone (PTH) PTH‒related peptide receptor is associated with abrupt cardiomyocyte death. Endocrinol. 2003;144(3):1053‒1061.
  72. Halapas A, Tenta R, Pantos C, et al. Parathyroid hormone‒related peptide and cardiovascular system. In Vivo. 2003;17(5):425‒432.
  73. Plawner LL, Philbrick WM, Burtis WJ, et al. Cell type specific secretion of parathyroid hormone‒related protein via the regulated versus the constitutive secretory pathway. J Biol Chem. 1995;270(23):14078‒14084.
  74. Vasavada RC, Cavaliere C, D'Ercole AJ, et al. Overexpression of parathyroid hormone‒related protein in the pancreatic islets of transgenic mice causes islet hyperplasia,hyperinsulinemia and hypoglycemia. J Biol Chem. 1996;271(2):1200‒1208.
  75. Vasavada RC, Wang L, Fujinaka Y, et al. Protein kinase C‒zetaactivation markedly enhances beta‒cell proliferation: An essential role in growth factor mediated beta‒cell mitogenesis. Diabetes. 2007;56(11):2732‒2743.
  76. Abraham P, Ralston SH, Hewison M, et al. Presentation of a PTHrP‒secreting pancreatic neuroendocrine tumor,with hypercalcemic crisis,pre‒eclampsia and renal failure. Postgrad Med J. 2002;78(926):752‒754.
  77. Bouvet M, Nardin SR, Burton DW, et al. Parathyroid hormone‒related protein as a novel tumor marker in pancreatic adenocarcinoma. Pancreas. 2002;24(3):284‒290.
  78. Kamp K, Feelders RA, van Adrichem RC, et al. Parathyroid Hormone‒Related Peptide (PTHrP) Secretion by Gastrenteropathic Neuroendocrine Tumors (GEP‒NETS): Clinical Features, Diagnosis, Management and Follow‒Up. J Clin Endocrinol Metab. 2014;99(9):3060‒3069.
  79. Weaver DR, Deeds JD, Lee K, et al. Localization of parathyroid hormone‒related peptide (PTHrP) and PTH/PTHrP receptor mRNAs in rat brain. Brain Res Mol Brain Res. 1995;28(2):296‒310.
  80. Weir EC, Brines ML, Ikeda K, et al. Parathyroid hormone‒related peptide gene is expressed in the mammalian central nervous system. Proc Natl Acad Sci USA. 1990;87(1):108‒112.
  81. Chatterjee O, Nakchbandi IA, Philbrick WM, et al. Endogenous parathyroid hormone‒related protein functions as a neuroprotective agent. Brain Res. 2002;930(1‒2):58‒66.
  82. Chattopadhyay N, Evliyaoglu C, Heese O, et al. Regulation of secretion of PTHrP by Ca(2+)‒sensing receptor in human astrocytes,astrocytomas and meningeomas. Am J Physiol Cell Physiol. 2000;297(3):691‒699.
  83. Funk JL, Trout CR, Wei H, et al. Parathyroid hormone‒related protein (PTHrP) induction in reactive astrocytes following brain injury: A possible mediator of CNS inflammation. Brain Res. 2001;915(2):195‒209.
  84. Shankar PP, Wei H, Davee SM, et al. Parathyroid hormone‒related protein is expressed by transformed and fetal human astrocytes and inhibits cell proliferation. Brain Res. 2000;868(2):230‒240.
  85. Pardo FS, Lien WW, Fox HS, et al Parathyroid hormone‒related expression is correlated with clinical course in patients with glial tumors. Cancer. 2004;101(11):2622‒2628.
  86. Mucica CM, Liang G Lankford KL, et al. Induction of parathyroid hormone‒related peptide following peripheral nerve injury: Role as a modulator of Schwann cell phenotype. Glia. 2006;53(6):637‒648.
  87. Dobolyi A, Dimitrov E, Palkovits M, et al. The Neuroendocrine Effects of the PTH‒2 Hormone Receptor. Front Endocrinol (Lausanne). 2012;3:121.
  88. Boabaid F, Berry JE, Koh AJ, et al. The role of parathyroid hormone‒related protein in the regulation of osteoclastogenesis by cement oblasts. J Periodontol. 2004;75(9):1247‒1254.
  89. Calvi LM, Shin HI, Knight MC, et al. Constitutively active PTH/PTHrP receptor in odontoblasts alters odontoblast and ameloblast function and maturation Mech Dev. 2004;121(2004):397‒408.
  90. Philbrick WM, Dreyer BE, Nakchbandi IA, et al. Parathyroid hormone‒related protein is required for tooth eruption. Proc Natl Acad Sci USA. 1998;95(20):11846‒11851.
  91. Singh RJ. Humoral Hypercalcemia of Malignancy: The Case for Parathyroid Hormone‒Related Protein, With Caveats. Cli Lab News. 2016.
  92. Shimuzu N, Dean T, Tsang J. Novel Parathyroid Hormone (PTH) Antagonists That Bind to the Juxtamembrane Portion of the PTH/PTH‒Related Protein Receptor. J Biol Chem. 2005;280(3):1797‒1807.
  93. Mirza A, Bedinger D, Hunt R, et al. Impacting Humoral Hypocalcaemia of Malignancy (HHM) and associated PTH1R‒mediated morbidities: Characterization of an Anti‒PTH1R antagonist monoclonal antibody to reverse hypocalcaemia. AACR Annual Meeting LB‒L306. 2015.
  94. Choppin A, Bedinger D, Hunt R, et al. A Novel Anti‒PTH1R Antagonist Monoclonal Antibody Reverses Hypercalcemia Induced byPTH or PTHrP: A potential Treatment of Primary Hyperparathyroidism and Humoral Hypocalcaemia of Malignancy. ENDO, Annual Meeting SAT 399, USA. 2017.
  95. Breitkopf T, Leung G, Yu M, et al. The basic science of hair biology: what are the causal mechanisms for the disordered hair follicle? Dermatol Clin. 2013;31(1):1‒19.
  96. Holick MF, Ray S, Chen TC, et al. A parathyroid hormone antagonist stimulates epidermal proliferation and hair growth in mice. Proc Natl Acad Sci USA. 1994;91(17):8014‒8016.
  97. Wysolmerski JJ, Broadus AE, Zhou J, et al. Overexpression of parathyroid hormone‒related protein in the skin of transgenic mice interferes with hair follicledevelopment. Proc Natl Acad Sci USA. 1994;91(3):1133‒1137.
  98. Peters EM, Foitzik K, Paus R, et al. A new strategy for modulating chemotherapy induced alopecia, using PTH/Pthrp receptor agonist and antagonist. J Invest Dermatol. 2001;117(2):173‒178.
  99. Masmoudi A, Chermi ZM, Marrekchi S, et al. Cowden syndrome. J Dermatol Case Rep. 2011;5(1):8‒13.
  100. Fuleihan GH, Rubeiz N. Dermatologic manifestations of parathyroid‒related disorders. Clin Dermatol. 2006;24(4):281‒288.
  101. Skrok A, Bednarczuk T, Skwarek A, et al. The Effect of Parathyroid Hormones on Hair Follicle Physiology:Implications for Treatment of Chemotherapy‒induced Alopecia. Skin Pharmacol Physiol. 2015;28(4):213‒225.
  102. Cheloha RW, Gellman SH, Vilardaga JP, et al. PTH receptor 1‒signalling: mechanistic insights and therapeutic prospects. Nat Rev Endocrinol. 2015;11(12):712‒724.
  103. Grey A, Mitnick MA, Shapses S, et al. Circulating levels of interleukin‒6 and tumor necrosis factor alpha are elevated in primary hyperparathyroidism and correlate with markers of bone resorption‒ a clinical research center study. J Clin Endocrinol Metab. 1996;81(10):3450‒3454.
  104. Takahashi S, Hakata M, Aiba K. Evaluation of circulating plasma cytokines in cancer patients with high plasma parathyroid hormone‒related protein levels. Endocr Rel Cancer. 2003;10:403‒407.
  105. Firkin F, Schneider H, Grill V. Parathyroid hormone‒related protein in hypercalcemia associated with hematological malignancy. Leukemia and Lymphoma. 1988;29(5‒6):499‒506.
  106. Guise TA, Yin JJ, Taylor SD, et al. Evidence for a causal role of parathyroid hormone‒related protein in the pathogenesis of human breast cancer mediated osteolysis. J Clin Invest. 1996;98(7):1544‒1549.
  107. Hofbauer LC, Neubauer A, Heufelder AE. Receptor activator of nuclear factor‒kappaB ligand and osteoprotogerin: potential implications for the pathogenesis and treatment of malignant bone disease. Cancer. 2001;92(3):460‒470.
  108. Kohli SS, Kohli VS. Role of RANKL‒RANK/osteoprotogerin molecular complex in bone remodelling and its immunopathologic implications. Indian J Endocrinol Metab. 2011;15(3):175‒181.
  109. Yoneda T, Tanaka S, Kenji H. Role of RANKL/RANK in primary and secondarry breast cancer. World J Ortop. 2013;4(4):178‒185.
  110. Lacey DL, Boyle WJ, Simonet WS, et al. Bench to bedside: elucidation of the OPG‒RANK‒RANKL pathway and the development of denosumab. Nat Rev Drug Discov. 2012;11(5):401‒419.
  111. Mould DR, Green B. Pharmokinetics and pharmacodynamics of monoclonal antibodies; concepts and lessons for drug development. Bio Drugs. 2010;24(1):23‒39.
  112. Baron R, Ferrari S, Russell RG. Denosumab and bisphosphonates; different mechanisms of action and effects. Bone. 2011;48(4):677‒692.
  113. Ashihara N, Nakajima K, Nakamura Y, et al. Denosumab is Effective for Controlling Serum Calcium Levels in Patients with Humoral Hypercalcemia of Malignancy Syndrome: A Case Report on Parathyroid Hormone‒Related Protein Producing Cholangiocarcinoma. Intern Med. 2016;55(23):3433‒3457.
  114. FDA Approval for Denosumab. NIH‒National Cancer Institute. 2013.
  115. Kir S, White JP, Kleiner S, et al. Tumor derived PTH‒related protein triggers adipose tissue browning and cancer cachexia. Nature. 2014;513(7516):100‒104.
  116. Economopoulos KP, Liu W. PTH/PTHrP Receptor Mediates Cachexia in Models of Kidney Failure and Cancer. Cell Metab. 2016;23(2):315‒323.
  117. NIH Directors Blog. Cancer Cachexia: Might Ths Molecule Hold the Key? 2011.
  118. Del Ferraro C, Grant M, Kockzywas M, et al. Management of Anorexia‒Cachexia in Late Stage Lung Cancer Patients. J Hosp Palliat Nurs. 2012;14(6).
  119. Porporato PE. Understanding cachexia as a cancer metabolism syndrome. Oncogenesis. 2016;5:e200.
Creative Commons Attribution License

©2017 Naafs. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.