Due casi di sindrome pseudo-Bartter in età pediatrica: quando sospettare una rara modalità d’esordio di fibrosi cistica

Abstract

La fibrosi cistica è una patologia multisistemica e le modalità di comparsa, l’entità dei sintomi e il decorso sono estremamente variabili. Un possibile esordio/complicanza della malattia è la sindrome pseudo-Bartter, caratterizzata da iponatriemia, disidratazione ipocloremica e alcalosi metabolica, in assenza di una patologia renale. Tale sindrome si manifesta più frequentemente nel primo anno di vita e ha un picco di insorgenza nel periodo estivo.
In questo articolo descriviamo due casi di esordio di fibrosi cistica con sindrome di pseudo-Bartter in età pediatrica. Fondamentale per l’inquadramento diagnostico è stato interrogarsi sulle possibili cause di alcalosi metabolica associata a iponatriemia. L’esperienza maturata con il primo caso è stata fondamentale per l’iter diagnostico del secondo caso.

Parole chiave: fibrosi cistica, sindrome pseudo-Bartter, alcalosi metabolica, iponatriemia, pediatria

Introduzione

La fibrosi cistica (FC) è una malattia genetica a trasmissione autosomica recessiva, caratterizzata dalla mutazione del gene che codifica per una proteina (CFTR: cystic fibrosis transmemebrane regulator) coinvolta nel trasporto transmembrana del cloro. Il gene responsabile si trova a livello del braccio lungo del cromosoma 7. Il CFTR è espresso sulle cellule epiteliali delle vie aeree, del tratto digerente, del pancreas, delle vie biliari, nelle ghiandole sudoripare e nell’apparato genitourinario. Le conseguenze del malfunzionamento sono l’incapacità di eliminare secrezioni mucose per lo scarso contenuto di acqua, un elevato contenuto di sali nel sudore e nelle altre secrezioni sierose e infezioni respiratorie croniche. La mutazione più comune del gene è una delezione che determina l’assenza di fenilalanina nella posizione 508 (ΔF508), tuttavia sono state descritte più di 2.000 mutazioni e le conseguenze molecolari di tali mutazioni possono essere raggruppate in 7 classi sulla base del tipo di alterazione (sintesi, maturazione, trasporto, funzionalità) [1]. 

La visualizzazione dell’intero documento è riservata a Soci attivi, devi essere registrato e aver eseguito la Login con utente e password.

L’inefficacia del tolvaptan correla con una prognosi negativa a breve termine nel paziente con tumore polmonare e sindrome da inappropriata antidiuresi

Abstract

Introduzione: Il tolvaptan (TVP), antagonista del recettore della vasopressina, rappresenta una opzione terapeutica della sindrome da inappropriata antidiuresi (SIAD). L’obiettivo di questo studio è stato quello di valutare gli effetti del TVP in pazienti oncologici con iponatriemia.
Metodi: Sono stati arruolati 15 pazienti oncologici affetti da SIAD. I pazienti trattati con TVP appartenevano al gruppo A, mentre pazienti iponatriemici trattati con soluzioni ipertoniche e restrizione di fluidi costituivano il gruppo B.
Risultati: Nel gruppo A, la correzione ottimale del sodio è stata ottenuta dopo 3.7±2.8 giorni. Nel gruppo B, i livelli target di sodiemia sono stati raggiunti più lentamente rispetto al gruppo A, dopo 5.2±3.1 giorni. La durata della degenza e l’incidenza di riospedalizzazione era maggiore nel gruppo B rispetto al gruppo A. Nel gruppo A, il 37% dei pazienti trattati con TVP ha avuto una recidiva di iponatriemia, nonostante il progressivo incremento della posologia del farmaco, da 7,5 mg sino a 60 mg die, con una completa mancanza di risposta al farmaco. In tali pazienti si è documentata una progressione di malattia, definita come crescita tumorale o comparsa di nuove lesioni metastatiche.
Conclusioni: Il Tolvaptan corregge l’iponatriemia correlata a una SIAD in modo più efficace e duraturo rispetto alle soluzioni ipertoniche e alla restrizione di liquidi. Tale efficacia determina un maggior numero di cicli di chemioterapia portati a termine, una minore durata della degenza, un minor tasso di recidiva dell’iponatriemia e della riospedalizzazione. Una mancata risposta al tolvaptan deve porre il sospetto di una progressione della malattia oncologica.

Parole chiave: Iponatriemia, sindrome paraneoplastica, sindrome da inappropriata antidiuresi, tolvaptan, vasopressina

Ci spiace, ma questo articolo è disponibile soltanto in inglese.

Introduction

The prognosis of oncologic patients is often related to the onset of electrolytic disorders, particularly if hyponatremia occurs [1]. The syndrome of inappropriate anti-diuresis (SIAD) represents the main cause of hyponatremia, even though differential diagnosis with concomitant comorbidities (heart failure, nephrotic syndrome, extracellular volume depletion, pulmonary disorders) and drugs (tricyclic antidepressants, selective serotonin reuptake inhibitors, opioids, chemotherapeutic agents and immunotherapy) needs to be carried out [2, 3].

In particular, SIAD is directly associated with malignancy as expression of a paraneoplastic endocrine effect mediated by an ectopic production of vasopressin (AVP) by cancer cells. Moreover, medications, particularly chemotherapic agents, such as vinca alkaloids, alkylating agents, and platinum compounds, which increase the AVP synthesis/release, could induce SIAD. Other drugs, such as cyclophosphamide, could enhance the water permeability of the distal tubule, in the absence of high AVP levels [1].

Fluid restriction remains the mainstay of treatment for acute and moderate hyponatremia associated with SIAD but inefficacy and frequent side effects, such as dehydration and acute kidney injury, often involving patients treated, at the same time with chemotherapy, require pharmacological interventions [4]. Demeclocycline, loop diuretics associated with sodium supplementation, and urea tablets could correct and solve hyponatremia [5]. In particular, the infusion of hypertonic saline (3%) is highly recommended in acute situations with neurological symptoms. The guidelines advise a bolus of 100–150 mL in 10 minutes, which might be repeated 2 to 3 times until serum sodium increase by 5 mmol avoiding overcorrection [6].
No more than 10 mmol in the first 24 hours or 8 mmol if there are risk factors must be reached in order to prevent severe damage to the central nervous system, such as central pontine myelinolysis, ultimately coma and death. The recommendation is to carry on the correction until symptoms’ disappearance, with careful monitoring of patient’s conditions and serum sodium concentration to avoid hyponatremia overcorrection. If the patient is symptomatic but hyponatremia occurred chronically, correction should be performed more gradually (1.5 to 2 mmol/L/h) [7].
Tolvaptan (TVP), an AVP receptor antagonist, represents a therapeutic option in these patients. It induces a net increase in free water excretion, decreasing aquaporin-2 channels in the renal collecting tubules and water re-absorption and consequently increasing serum sodium concentrations. This phenomenon causes a pure aquaresis without a rise of sodium or potassium urinary excretion [8].
Various studies have investigated the usefulness of TVP, revealing its safety and efficacy in hyponatremic patients, with additional benefits in terms of quality of life and reduced hospitalization [9, 10]. The correction of hyponatremia may also improve anti-neoplastic effects of chemotherapies [11], such as the decrease of AVP levels could reflect a neoplastic remission, or conversely, a recurrence [12].
Despite these findings, hyponatremia did not correlate with tumor burden. Although several studies have focused on hyponatremia in cancer patients, only a limited number of case reports of SIAD are available in this setting, and a limited amount of data on SIAD in cancer patients are available in the literature.
This observational, hypothesis generating study aimed to evaluate the efficacy and safety of TVP to treat and solve hyponatremia in oncologic patients with SIAD. Moreover, we compared TVP results with those obtained by intravenous (iv) therapy based on hypertonic solutions. The incidence of re-hospitalization, due to hyponatremia recurrence and consequent prognostic implication of TVP resistance was evaluated.


Patients and methods

15 adult patients, with a histologically confirmed diagnosis of solid pulmonary tumors who have developed SIAD between January 2017 and June 2020, were retrospectively enrolled in the study.
Diagnostic criteria confirmed the SIAD [13] by evaluating the volemic status, serum sodium concentration and serum osmolality, urine sodium concentration and urine osmolality, thyroid function tests, and serum cortisol. Hypovolemic and hypervolemic hyponatremia has been excluded, as well as hyponatremia due to other endocrine causes, including adrenal insufficiency or hypothyroidism.
Patients with transient hyponatremia due to drugs (i.e., antidepressants, anticonvulsants, antipsychotic), who did not feel thirsty or have difficulty drinking water, with urinary tract obstruction, with serum sodium levels <115 mEq/L associated with severe neurologic deficits, such as seizures, have been excluded from the study. Furthermore, heart failure, renal failure with a glomerular filtration rate < 60 ml/min, ascites associated with hepatic cirrhosis, nephrotic syndrome, severe arterial hypotension, evidence of urinary tract obstruction, poorly controlled diabetes mellitus, a recent history of myocardial infarction, and cerebrovascular disorders determined the exclusion from the enrolment.

Hyponatremia correction and follow-up

According to the pharmacological strategy, patients receiving TVP belonged to group A, whereas group B was characterized by hyponatremic patients treated with hypertonic saline solutions and fluid restriction. In particular, group A patients were treated with 7.5 mg/day of TVP on Day 1 and increased to 15 mg/day if no serum sodium increase was revealed. Conversely, an infusion of 1 ml/kg/h of 3% hypertonic saline was administered in group B, with serum sodium checked every 2 hours during the first 12 hours of treatment.

Patients were discharged and managed as outpatients if they met ambulatory criteria, once the maintenance dose of TVP had been determined. In particular, after the hospital discharge, all patients have been followed through a dedicated outpatient clinic system, with different timing according to clinical conditions and sodium levels. “Maintenance criteria” required weekly serum sodium evaluation before TVP administration, with a target of 2 mEq/L of sodium difference if compared with the concentration assessed at the hospital discharge or the last serum assessment. The liver function panel has been also monitored, as well as body weight and vital signs. Moreover, to avoid rapid and potentially dangerous sodium concentration increase, group A patients did not have a fluid restriction.

The current study was undertaken following the principles outlined in the Declaration of Helsinki. Written informed consent was obtained from all patients. Clinical data were collected from medical chart reviews and electronic records.

Statistical analysis

Statistical analyses were performed with NCSS for Windows (version 4.0), the MedCalc (version 11.0; MedCalc Software Acacialaan, Ostend, Belgium) software, and the GraphPad Prism (version 5.0; GraphPad Software, Inc., San Diego, CA, USA) package. Data were presented as mean ± SD for normally distributed values (at Kolmogorov-Smirnov test) and median [IQ range] for not normally distributed values. Differences between groups were established by unpaired t-test or by ANOVA followed by Bonferroni’s test for normally distributed values and by Kruskal-Wallis analysis followed by Dunn’s test for nonparametric values. Dichotomized values were compared using the χ2 test. All results were considered significant if p was < 0.05.


Results

Patients baseline characteristics

Total Group A (TVP) Group B (Hypertonic) p
Patients 15 8 7 p > 0.05
Age, y 69.7±5.9 68.6±6.8 67.4±5.2 p > 0.05
WBC, mm3 6.07±2.4 5.24±2.8 6.37±1.9 p > 0.05
eGFR 78.4±12.8 81.2±9.7 77.4±10.1 p > 0.05
Admission [Na], mmol/l 120.4±2.9 119.7±3.8 118.4±2.9 p > 0.05
Admission Serum Osmolality, mOsm/Kg 257±12.3 244.2±9.3 247±11.2 p > 0.05
Admission Urine Osmolality, mOsm/Kg 407±119.8 465±79.2 397±101 p > 0.05
Hospital stay, days 17.9±8.2 17.6±4.7 11.2±3.4 p < 0.01
Δ[Na], mmol/l/24h 7.9±4.4 8.7±3.5 7.4±4.2 p > 0.05
[Na] Target Achievement, days 6.4±3.6 3.7±2.8 5.2±3.1 p: 0.01
[Na] Hospital Discharge 143.8±1.8 143.4±2.6 144.2±2.1 p > 0.05
Hyponatremia recurrence
Outpatients management, days 22.6±10.8 26.5±6.4 11±6.4 p < 0.01
Re-hospitalization, n (%) 8 (53) 3 (37) 5 (72) p > 0.05
[Na] re-admission, mmol/l 122.7±2.8 123.9±3.4 121.4±3.7 p:0.03
Re-admission Serum Osmolality 251±9.4 262±7.9 258±4.5 p > 0.05
Re-admission urine Osmolality 504±104.8 472±93.4 408.5±79 p > 0.05
Disease progression, n (%) 5 (62) 3 (100) 2 (40) p > 0.05
Table 1: Baseline characteristics of the study cohort.

A total of 15 patients (mean age: 69.7±5.9 years) were hospitalized because of SIAD, and the median sodium level at admission was 120.4±2.9 mmol/l (range 117.4-123.2 mmol/l). The median duration of hospitalization was 17.9±8.2 days (range 9.4-22.7 days). All patients underwent chest radiograph evaluation after the admission to the hospital, excluding active findings of pulmonary infection. The mean white blood cell count was 6.07±2.4 mm3.

Hyponatremia management

Eight patients received tolvaptan for SIAD treatment (group A), whereas hypertonic solutions and fluid restriction have been the treatment for the remaining seven patients (group B).

In group A, all patients started tolvaptan at a dose of 7.5 mg/daily, with the next modifications according to sodium and osmolarity levels.

Hyponatremia has been improved in all Group A patients, without toxicity due to the drug, as revealed by normal values of liver function tests.

Serum sodium levels have been monitored over the first 24 h at regular intervals of 4-6 h to check the correction speed. A rapid correction was not observed, with a slow improvement of sodium levels within the range of 8-10 mmol/l/24h (ΔNa: 8.7±3.5 mmol/l/24h). Moreover, the correction of serum sodium, defined at 135 mmol/l, was achieved after 3.7±2.8 days. All patients required a final dose of 7 or 15 mg of TVP, before hospital discharge, as chronic therapy (Figure 1).

Figure 1: Sodium trend during tolvaptan administration.
Figure 1: Sodium trend during tolvaptan administration.

In group B, patients had a similar trend for sodium increase (ΔNa: 7.4±4.2 mmol/l/24h), but the target levels were obtained more slowly, after 5.2±3.1 days (p: 0.01) than group A. Similarly, the hospital stay was longer in group B than in group A (17.6±4.7 vs 11.2±3.4 days; p< 0.01).

Hyponatremia relapse and re-hospitalization

Serum sodium levels were similar between the two groups during the last assessment in the hospital (143.4±2.6 vs 144.2±2.1; p> 0.05). After discharge, all patients have been followed weekly, to monitor clinical conditions and sodium levels.

Group B was characterized by a higher incidence of re-hospitalization than group A. In particular, 5/7 patients (72%) required hospitalization within 11±6.4 days after the discharge, due to symptomatic hyponatremia (mean serum sodium value at the hospital admission was 121.4±3.7 mmol/l), determining drowsiness, mental confusion or fatigue. This condition was solved by repeating intravenous schemes with hypertonic solutions, with a mean hospital stay of 13.6 ±7.2 days.

In TVP group A, only 3/8 patients (37%) had hyponatremic relapses. These three patients were managed in the outpatient clinic after 26.5±6.4 days (vs 11±6.4 days; p < 0.01) since the last hospital discharge. During this period, serum sodium was maintained between 140 and 145 mmol/l, administering 7.5 or 15 mg of TVP, daily or every other day. In these three patients, severe, sudden and detrimental hyponatremia was observed (123.9±3.4 mmol/l), requiring re-hospitalization. Notwithstanding the progressive increase of doses of TVP, from 7.5 or 15 mg to 60 mg per day hyponatremia did not improve, revealing a complete lack of response to TVP.

Behind planned protocols, the unscheduled oncologic and radiologic staging was early performed. In all three patients, a growth of tumor mass or new metastatic lesions has been revealed, requiring a re-scheduling and modification of chemotherapy cycles. Only after some chemotherapy administration, sodium levels slowly increased. Figure 2 synthetizes the events occurred in a patient with TVP resistance.  

Figure 2: Management of hyponatremia and oncologic restaging in a patient with tolvaptan resistance.
Figure 2: Management of hyponatremia and oncologic restaging in a patient with tolvaptan resistance.

In the remaining five patients (63%) of group A, hyponatremic episodes were often acutely and temporarily related to chemotherapy administration, with sodium correction after adjustment of TVP dose. These patients have been managed in outpatients clinic for more than six months, without the requirement of re-hospitalization and with no progression of cancer disease revealed after scheduled follow-up. All patients completed the planned chemotherapy.

No patients discontinued the TVP treatment due to adverse events, liver dysfunction, or serum sodium concentration exceeding 145 mEq/L.


Discussion

TVP represents a safe and valid therapeutic option to treat hyponatremic oncologic patients who developed SIAD, with no rapid sodium increase observed during the study period and without side effects TVP-related, such as sodium overcorrection or hepatic toxicity. Moreover, the aquaretic approach immediately increased serum sodium concentration, reducing the length of hospitalization, if compared to alternative methods, such as hypertonic solutions, reaching sodium normalization, but in fewer patients and in a longer time.

TVP also reduced the incidence of relapse of hyponatremia and re-hospitalization rate in outpatient follow-up, with obvious positive consequences on quality of life and costs.

These positive data also derived from specific clinical management of these patients, requiring multidisciplinary approaches and strategies, through a dedicated outpatient clinic system after the hospital discharge. Our patients have been evaluated with different timing, according to sodium levels and, consequently, TVP dosage adjustment.

A specific focus on these patients determined various supplementary effects, behind the issue “natremia”. During the follow-up, no TVP patients experienced acute kidney injury in concomitance with chemotherapeutical cycles, probably due to the diminished incidence of dehydration, secondary to fluid restriction, which could be usually prescribed in patients with low sodium levels. The maintenance of serum sodium within the normal range allowed the precocious start of chemotherapeutic agents, without chemotherapy-related hyponatremia, due to the intravenous fluid overload and drug-associated AVP secretion. Behind these positive effects, related to the quick start of chemotherapy, TVP treatment facilitated the administration of chemotherapy cycles constantly and promptly, avoiding delays. Moreover, the efficacy of anticancer treatment is notably improved in patients with normal sodium levels or after its correction, as recently stated by Berardi who revealed an optimal sodium correction, an improvement of the hospitalization length and quality of life during TVP treatment [14].

Our study also suggested potential prognostic information that could be deduced from TVP patients, in whom sudden and progressive hyponatremia occurred, despite TVP dosage increase. A re-staging of these patients highlighted tumor mass growth or new metastatic lesions, suggesting cancer AVP production, requiring modification of chemotherapy cycles.

However, notwithstanding all this available evidence, the use of TVP remains limited in daily clinical practice, around 5%, according to the hyponatremia registry [15].

Adequate management of natremia is crucial due to the well-known correlation with overall survival, both in the general population and in cancer patients. Many studies revealed a close link between low sodium levels and poor prognosis, not only in small cell lung cancer [14] but also in other types of non-pulmonary tumors, including renal cell carcinoma and gastrointestinal cancer [1619].

We cannot analyze potential relationships between survival and sodium correction, acutely or during a long follow-up period, by TVP administration due to the small cohort of patients.

The present study has some limitations that should be mentioned. First, the retrospective nature of this study could result in unwanted methodological biases. The therapy choices were not randomized; therefore, conclusions about the relative efficacy of the treatments, including TVP, are limited. Confirmation in wider cohorts is indispensable to attribute general validity to our results.

These limitations did not allow us to strengthen the prognostic role of TVP failure to improve sudden hyponatremia or to evaluate the impact of TVP therapy and sodium maintenance on patients’ survival. However, our study generating hypotheses could be a starting point for further studies.

 

Conclusions

Our data demonstrated that TVP improved hyponatremia more efficiently and stably than hypertonic solutions and fluid restrictions, with a high rate of chemotherapeutical cycles concluded. Positive consequences have been obtained about the hospital stay, rate of relapse of hyponatremia, and re-hospitalization. TVP exerted its potential benefit of long-term use in an outpatient setting, improving the quality of life of SIAD oncologic patients.

 

Acknowledgments

The authors acknowledge the Nephrology nurses Carmelo Saterno and Caterina Ragno for their clinical work during the outpatient clinic management and for participating in the study collecting data.

 

Bibliography

  1. Workeneh BT, Jhaveri KD, Rondon-Berrios H. Hyponatremia in the cancer patient. Kidney Int 2020; 98:870 – 882. https://doi.org/1016/j.kint.2020.05.015.
  2. Doshi SM, Shah P, Lei X et al. Hyponatremia in hospitalized cancer patients and its impact on clinical outcomes. Am J Kidney Dis 2012; 59:222–228. https://doi.org/1053/j.ajkd.2011.08.029.
  3. Lacquaniti A, Altavilla G, Picone A, et al. Apelin beyond kidney failure and hyponatremia: a useful biomarker for cancer disease progression evaluation. Clin Exp Med 2015; 15:97-105. https://doi.org/1007/s10238-014-0272-y.
  4. Thajudeen B and Salahudeen AK. Role of tolvaptan in the management of hyponatremia in patients with lung and other cancers: current data and future perspectives. Cancer Manag Res 2016; 8: 105–114. https://doi.org/2147/CMAR.S90169.
  5. Cawley MJ. Hyponatremia: current treatment strategies and the role of vasopressin antagonists. Ann Pharmacother 2007; 41:840–50. https://doi.org/1345/aph.1H502.
  6. Spasovski G, Vanholder R, Allolio B, et al. Clinical practice guideline on diagnosis and treatment of hyponatraemia. Nephrol Dial Transplant 2014; 29:1–39. https://doi.org/1530/EJE-13-1020.
  7. Verbalis JG, Adler S, Schrier RW, Berl T, Zhao Q, Czerwiec FS. SALT Investigators. Efficacy and safety of oral tolvaptan therapy in patients with the syndrome of inappropriate antidiuretic hormone secretion. Eur J Endocrinol 2011;164:725–732. https://doi.org/1530/EJE-10-1078.
  8. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2799145/Reilly T, Chavez B. Tolvaptan (Samsca) for Hyponatremia: is it worth its Salt? Pharmacy and Therapeutics 2009; 34:543–547.
  9. Jamookeeah C, Robinson P, O’Reilly K et al. Cost-effectiveness of tolvaptan for the treatment of hyponatraemia secondary to syndrome of inappropriate antidiuretic hormone secretion in Sweden. BMC Endocr Disord 2016; 16:22. https://doi.org/1186/s12902-016-0104-z.
  10. Fiordoliva I, Meletani T, Baleani MG et al. Managing hyponatremia in lung cancer: latest evidence and clinical implications. Ther Adv Med Oncol 2017; 9:711-719. https://doi.org/1177/1758834017736210.
  11. Doshi KH, Shriyan B, Nookala MK et al. Prognostic significance of pretreatment sodium levels in patients of nonsmall cell lung cancer treated with pemetrexed-platinum doublet chemotherapy. J Cancer Res Ther 2018; 14:1049–1053. https://doi.org/4103/0973-1482.187296.
  12. Kanaji N, Watanabe N, Kita N et al. Paraneoplastic syndromes associated with lung cancer. World J Clin Oncol 2014; 5:197-223. https://doi.org/5306/wjco.v5.i3.197.
  13. Verbalis JG, Goldsmith SR, Greenberg A et al. Diagnosis, evaluation, and treatment of hyponatremia: expert panel recommendations. Am J Med 2013; 126: S1–42. https://doi.org/1016/j.amjmed.2013.07.006.
  14. https://pubmed.ncbi.nlm.nih.gov/31632468/Berardi R, Mastroianni C, Lo Russo G et al. Syndrome of inappropriate anti-diuretic hormone secretion in cancer patients: results of the first multicenter Italian study. Ther Adv Med Oncol 2019; 11:1– 13. https://doi.org/1177/1758835919877725.
  15. Greenberg A, Verbalis JG, Amin AN et al. Current treatment practice and outcomes. Report of the hyponatremia registry. Kidney Int 2015; 88:167–77. https://doi.org/1038/ki.2015.4.
  16. Tiseo M, Buti S, Boni L et al. Prognostic role of hyponatremia in 564 small cell lung cancer patients treated with topotecan. Lung Cancer 2014; 86: 91–95. https://doi.org/1016/j.lungcan.2014.07.022.
  17. Farid SG and Prasad KR. Prognostic impact of hyponatraemia in patients with colorectal cancer. Colorectal Dis 2015; 17: 451. https://doi.org/10.1111/codi.12939.
  18. Jeppesen AN, Jensen HK, Donskov F et al. Hyponatremia as a prognostic and predictive factor in metastatic renal cell carcinoma. Br J Cancer 2010;102: 867–872. https://doi.org/1038/sj.bjc.6605563.
  19. Bolignano D, Medici MA, Coppolino G et al. Aquaretic inhibits renal cancer proliferation: Role of vasopressin receptor-2 (V2-R). Urologic oncology 2010; 28:642–647. https://doi.org/1016/j.urolonc.2008.12.014.

La “pathway” della Vasopressina come target farmacologico in ambito nefrologico: una review narrativa

Abstract

L’ADH è un ormone secreto dalla neuroipofisi che svolge diversi ruoli in base all’organo bersaglio. A livello renale questo peptide è in grado di causare assorbimento di acqua libera da elettroliti giocando pertanto un ruolo fondamentale nell’equilibrio idro-elettrolitico. Esistono patologie e disturbi che mettono a repentaglio tale bilanciamento e, in questo campo, farmaci inibitori recettoriali dell’ADH come i vaptani potrebbero svolgere un ruolo fondamentale. Inibendo la via di attivazione della vasopressina, teoricamente trovano indicazione nelle iponatriemie ipotoniche ipervolemiche ed euvolemiche. Tuttavia i trial clinici nello scompenso cardiaco non hanno dato risultati favorevoli su outcome clinici. Anche nella SIADH, nonostante il loro largo impiego, le evidenze in favore dei vaptani sono tali che non vi è accordo tra gli esperti circa il loro impiego.

Poiché i vaptani inibiscono la pathway dell’AMPc nelle cellule tubulari, è stato proposto il loro impiego per inibire la cistogenesi. Un trial clinico ha dimostrato un effetto favorevole sulla progressione dell’ADPKD.

In considerazione degli effetti dell’ADH in modelli di patologie cistiche renali differenti dall’ADPKD, è stato suggerito l’impiego dei vaptani in patologie quali la nefronoftisi e la malattia policistica autosomica recessiva. Altri possibili impieghi potrebbero essere in ambito trapiantologico renale e nella sindrome cardio-renale.

Per il ruolo di ADH nei meccanismi coagulativi ed emostatici, sfruttando la via di attivazione dell’ADH attraverso i suoi analoghi come la Desmopressina Acetato, si può ridurre il rischio di sanguinamento nelle biopsie in pazienti a rischio emorragico.

Parole chiave: vasopressina, vaptani, iponatriemia, ADPKD, biopsia

Introduzione

La vasopressina, nota anche come adiuretina o arginin-vasopressina (AVP) o ormone antidiuretico (ADH), è un ormone neuropeptidico prodotto a livello dei nuclei sopraottico e paraventricolare che viene secreto dall’ipofisi posteriore in risposta ad un aumento della tonicità plasmatica o alla diminuzione del volume plasmatico (1).

 

La visualizzazione dell’intero documento è riservata a Soci attivi, devi essere registrato e aver eseguito la Login con utente e password.