Correlation between hypophosphatemia and renal diseases and the progress in diagnosis and treatment

Zhang Biyu, Chen Nan

PDF(3892 KB)
Chinese Journal of Nephrology ›› 2020, Vol. 36 ›› Issue (9) : 716-725. DOI: 10.3760/cma.j.cn441217-20200330-00128
Review

Correlation between hypophosphatemia and renal diseases and the progress in diagnosis and treatment

Author information +
History +

Abstract

Cite this article

Download Citations
Zhang Biyu. , Chen Nan. Correlation between hypophosphatemia and renal diseases and the progress in diagnosis and treatment[J]. Chinese Journal of Nephrology, 2020, 36(9): 716-725. DOI: 10.3760/cma.j.cn441217-20200330-00128.
磷作为机体的生命必需元素,在细胞新陈代谢、信号转导、蛋白合成、参与细胞膜及骨质构成等方面发挥着重要作用,而肾脏是调节磷代谢平衡最为重要的脏器。低磷血症(hypophosphatemia)与高磷血症(hyperphosphatemia)在肾脏病中均较为常见。慢性肾脏病(chronic kidney disease,CKD)患者因肾小球滤过率(glomerular filtration rate,GFR)下降,CKD 3b期后常出现高磷血症[1],进而引起以矿物质和内分泌代谢紊乱、骨骼异常、血管及其他软组织钙化为主要特点的综合征,即慢性肾脏病矿物质与骨异常(chronic kidney disease-mineral and bone disorder,CKD-MBD)。目前改善全球肾脏病预后组织(Kidney Disease:Improving Global Outcomes,KDIGO)与中国国家肾脏疾病临床医学研究中心均已制定CKD-MBD诊治指南[2-4],因此在高磷血症的认识与诊疗方面已形成较为完善的体系。相比之下,低磷血症与肾脏病的关联虽十分密切,相关研究也已取得一定进展,但临床医务工作者普遍缺乏系统性认知,其治疗方面更是存在很多漏洞。
低磷血症在人群中的发病率鲜有报道,国内仅有个别医院对住院患者血磷水平进行统计,其中低磷血症患者占总住院患者的6.5%[5],而在重症监护室中低磷血症发病率可高达40.95%[6],低磷血症与住院患者病死率及重症监护室患者的预后密切相关。各种原因导致肾脏对于尿磷的重吸收减少均会导致低磷血症,进而引起骨骼、肌肉、神经等系统的异常表现。本综述着眼于低磷血症与肾脏病的关联,同时对低磷血症的诊治进展做详细阐述,以期提高肾脏科临床医务工作者对低磷血症的系统性认识,熟悉诊疗思路。

一、 血磷的稳态

正常成年人每日约经肠道吸收0.9~1.0 g的磷元素,近端小肠为吸收的主要部位,吸收形式主要为无机磷酸盐(H2PO4-或HPO42-[7-8]。吸收入体内的磷一部分构成骨质,促进骨基质形成及矿化,另一部分存在于组织中,用于体内新陈代谢,参与核酸、ATP、酶及蛋白质的合成与调控。机体内的磷主要通过肾脏排泄,血磷可经肾小球全部滤过,近80%的尿磷可被肾近端小管重吸收[7,9-10],肠道亦可排磷。在正常人体内,骨是贮存磷的主要部位,存储约80%~90%的磷元素,组织则包含了10%~20%的磷元素,血磷总量仅占体内磷元素总量的2%~3%[11-12],其稳态调节见图1。血磷的正常值范围因年龄而异,正常成年人血磷的参考范围为0.8~1.45 mmol/L[7],未成年儿童及青少年的血磷参考范围随着年龄增加而降低[13],详见表1
图1 血磷稳态调节示意图
注:(-):降低;(+):升高;FGF23:成纤维细胞生长因子23;PTH:甲状旁腺素

Full size|PPT slide

表1 不同年龄段血磷正常值参考范围
年龄段 血磷参考范围(mmol/L)
0~5 d 1.55~2.65
1~3岁 1.23~2.10
4~11岁 1.19~1.81
12~15岁 0.84~1.74
16~19岁 0.87~1.52
正常成年人 0.80~1.45
磷的吸收与重吸收主要通过分布于小肠及肾近曲小管刷状缘的钠磷共转运体(sodium phosphate cotransporter,NPT)完成,该过程主要为主动运输[8]。人体内的钠磷共转运体共有3种类型,其中2型钠磷共转运体(NPT2)为负责体内吸收磷的主要转运体,其亦分3种类型:NPT2a与NPT2c主要分布于肾近曲小管,重吸收尿磷;NPT2b主要分布于小肠,负责肠道对磷的吸收。其余转运体,如Pit-2等也参与磷元素的转运,但其在维持体内磷稳态的重要性仍待进一步验证[11,14]
体内调控磷代谢的激素主要有3种:活性维生素D[即骨化三醇,1,25-(OH)2-D3]、甲状旁腺素(parathyroid hormone,PTH)、成纤维细胞生长因子23(fibroblast growth factor 23,FGF23)。维生素D(骨化醇)经肝脏25-羟化酶及肾脏1α-羟化酶羟化转变为活性维生素D,作用于维生素D受体,继而上调NPT2的表达,促进磷的吸收与重吸收以升高血磷。PTH是由甲状旁腺分泌的碱性单链多肽类激素,下调NPT2的表达,抑制肾小管及肠道对磷的(重)吸收,使得血磷降低。FGF23主要由骨组织产生,作用于其受体FGFR,在共受体α-klotho的帮助下,下调NPT2表达以降低血磷。三种激素与血磷之间存在相互影响。低血磷可促进活性维生素D产生以升高血磷,高血磷则促进PTH产生以降低血磷,活性维生素D可刺激FGF23表达、抑制PTH合成,而PTH可促进活性维生素D合成,FGF23则抑制活性维生素D合成、促使其分解[14-15]。这三种调磷激素均可作用于肾脏,通过控制NPT2的表达维持血磷的稳定。因此,无论是NPT2本身的异常或是调磷激素的异常均可导致肾脏排磷过多,从而引起原发性或继发性肾脏相关低磷血症,以下简称为肾性低磷血症。

二、 肾性低磷血症的病因分类

肾性低磷血症根据其病因为NPT/肾小管本身功能异常或是调磷激素异常可分为原发性和继发性两大类,根据遗传方式又可分为遗传性和获得性两大类,此处将按照病因进行分类与阐述,同时按照遗传方式分类列表详细描述其特征,获得性肾性低磷血症详见表2,遗传性肾性低磷血症详见表3。由于遗传性低磷血症常自幼儿及儿童期发病,大多伴随佝偻病(rickets)表现,故下文不再区分遗传性低磷血症与遗传性佝偻病。
表2 获得性肾性低磷血症分类、致病机制及特征
疾病类型 致病机制 血磷 血钙 尿磷 尿钙 TmP/GFR FGF23 25-(OH)-D3 1,25-(OH)2-D3 PTH AKP 其余信息
获得性维生素D
相关型肾性低磷
血症
维生素D摄入不足 N/↓ ↑/N/↓ N ↑↑ ↑↑
药物诱导P450致活性维生素D
分解增加
N/↓

↑/N/↓


N
N/↓

↑↑
↑↑
相关药物:苯妥英、苯巴比妥、卡马西平、异烟肼、利福平
获得性FGF23相关
型肾性低磷血症
肿瘤性骨软化症 N/↓ N N/↓ N/↑
药物引起FGF23升高

N/↓




N
N/↓
N/↑

相关药物:静脉麦芽糖铁、核苷酸类抗病毒药
获得性PTH相关型
肾性低磷血症

原发性甲状旁腺功能亢进或
药物直接引起PTH升高
N/↓
↑/N
↑/N/↓


N
N

↑↑
↑↑
相关药物:特立帕肽
钙摄入缺乏或药物引起低血钙
N/↓

↑/N/↓


N
N

↑↑
↑↑
相关药物:帕米膦酸钠、唑来膦酸、地诺单抗、酪氨酸激酶抑制剂
获得性NPT相关型
肾性低磷血症
药物引起肾脏近端小管NPT2
表达下调

N




N

N/↓

相关药物:阿昔洛韦、雌激素
获得性非NPT相关
型肾小管损伤导致
的肾性低磷血症
药物引起小管上皮细胞线粒体
损伤、基底膜Na+/K+ATP酶活性
降低、直接肾毒性



N/↓





N/↑





↓/N


N


N/↑


↑/N/↓





相关药物:酪氨酸激酶抑制剂、丙戊酸、利福平、多种化疗药、氨基糖苷类抗生素、核苷类抗病毒药、蛋白酶抑制剂、非甾体类抗炎药
注:N:正常;↑:升高;↑↑:明显升高;↓:降低;TmP/GFR:最大肾小管磷重吸收率与肾小球滤过率之比值;FGF23:成纤维细胞生长因子23;PTH:甲状旁腺素;AKP:碱性磷酸酶;NPT:钠磷共转运体
表3 遗传性肾性低磷血症分类、致病机制及特征
疾病类型 疾病名称
(英文缩写)
基因 位置 遗传
方式
致病机制 血磷 血钙 尿磷 尿钙 TmP/GFR FGF23 25-(OH)-D3 1,25-(OH)2-D3 PTH AKP 其余特点
维生素D相关型遗传性肾性低磷血症 维生素D依赖型佝偻病1A(VDDR1A) CYP27B1 12q14.1 AR 1α-羟化酶功能缺陷 N/↓ ↑/N/↓ ↓/N N ↑↑ ↑↑
维生素D依赖型佝偻病1B(VDDR1B) CYP2R1 11p15.2 AR 25-羟化酶功能缺陷 N/↓ ↑/N/↓ ↓/N ↓/N ↑↑ ↑↑
维生素D依赖型佝偻病2A(VDDR2A) VDR 12q13.11 AR 维生素D受体功能缺陷 N/↓ ↑/N/↓ ↓/N N ↑↑ ↑↑ 秃顶
维生素D依赖型佝偻病2B(VDDR2B) HNRNPC 14q11.2 ? 过度表达某种核糖核蛋白与VDR竞争结合维生素D受体元件 N/↓ ↑/N/↓ ↓/N N ↑↑ ↑↑
FGF23相关型遗传性肾性低磷血症 X连锁低磷血症(XLH) PHEX Xp22.11 XD FGF23分泌增加 N N/↓ N N/↑
常染色体显性低磷血症性佝偻病(ADHR) FGF23 12p13.32 AD FGF23抵抗降解 N N/↓ N N/↑
常染色体隐性低磷血症性佝偻病(ARHR1) DMP1 4q22.1 AR FGF23释放增多 N N/↓ N N/↑
常染色体隐性低磷血症性佝偻病(ARHR2) ENPP1 6q23.2 AR FGF23释放增多 N N/↓ N N/↑
Raine综合征(ARHR3) FAM20C 7p22.3 AR FGF23释放增多 N N/↓ N N/↑ 全身多脏器系统结构畸形及发育异常
骨挖空发育不良(OGD) FGFR1 8p11.23 AD FGF23受体功能增强 N N N N N N/↑ 全身多脏器系统结构畸形及发育异常
NPT相关型遗传性肾性低磷血症 低磷血症伴肾结石或骨质疏松1(NPHLOP1) SLC34A1 5q35.3 AD,AR NPT2a突变 N N N/↓ 肾结石
低磷血症伴肾结石或骨质疏松2(NPHLOP2) SLC9A3R1 17q25.1 AD NHERF1调节蛋白异常致NPT2a表达下调 N N N/↓ 肾结石
遗传性低磷血症性佝偻病伴高尿钙(HHRH) SLC34A3 9q34.3 AR NPT2c突变 N N N/↓ 肾结石
非NPT相关型肾小管损伤导致的肾性低磷血症 Dent病1型 CLCN5 Xp11.23 XLR 电压门控的氯离子通道5突变 N/↓ ↓/N N ↑/N/↓ 肾结石,中低分子蛋白尿,氨基酸尿,葡萄糖尿,血尿
Dent病2型 OCRL Xq26.1 XLR 细胞内吞异常 N/↓ ↓/N N ? ? 肾结石,中低分子蛋白尿,氨基酸尿,肌酸激酶及乳酸脱氢酶升高
范可尼肾小管综合征1型 FRTS1 15q15.3 AD ? N/↓ N/↑ N N N N 肾结石,中低分子蛋白尿,氨基酸尿,葡萄糖尿,肾性失钠失钾,乳酸尿
范可尼肾小管综合征2型 SLC34A1 5q35.3 AR ? N/↓ N N/↓ 肾结石,中低分子蛋白尿,氨基酸尿,葡萄糖尿,肾性失钠失钾,高尿酸尿
范可尼肾小管综合征3型 EHHADH 3q27.2 AD 线粒体呼吸及ATP合成障碍 N/↓ N/↑ N N N N 肾结石,中低分子蛋白尿,氨基酸尿,葡萄糖尿,肾性失钠失钾
范可尼肾小管综合征4型伴成年发作的青年型糖尿病 HNF4A 20q13.12 AD ? N/↓ N/↑ N N N N 肾结石,中低分子蛋白尿,氨基酸尿,葡萄糖尿,肾性失钠失钾,低尿酸血症
注:N:正常;↑:升高;↑↑:明显升高;↓:降低;TmP/GFR:最大肾小管磷重吸收率与肾小球滤过率之比值;FGF23:成纤维细胞生长因子23;PTH:甲状旁腺素;AKP:碱性磷酸酶;NPT:钠磷共转运体;NHERF1:Na+/H+交换调节因子1;AD:常染色体显性遗传;AR:常染色体隐性遗传;XD:伴X染色体显性遗传;XLR:X连锁隐性遗传;?:未明
1. 维生素D相关型肾性低磷血症: 此类肾性低磷血症是由于体内维生素D缺乏或功能异常所致,可进一步分为获得性和遗传性两类。获得性维生素D相关型肾性低磷血症系由各种原因导致活性维生素D水平降低,同时低血钙导致继发性甲状旁腺功能亢进,两者共同导致低磷血症的发生。其中维生素D摄入不足是获得性肾性低磷血症最常见的病因,可导致营养性佝偻病。此外,一些抗癫痫药和抗结核药可以诱导细胞色素P450导致活性维生素D分解增加,亦可导致低磷血症的发生[16],相关药物见表2
遗传性维生素D相关型肾性低磷血症可依据活性维生素D是否合成不足分为维生素D依赖型佝偻病(vitamin D dependent rickets,VDDR)和维生素D抵抗型佝偻病。前者通过影响活性维生素D合成所需的羟化酶导致低磷;后者在最初亦被称为VDDR,系维生素D受体突变或无法发挥正常功能[17]所致。各类型遗传性维生素D相关性佝偻病的具体信息及特点详见表3
2. FGF23相关型肾性低磷血症: 该类型肾性低磷血症是由于体内FGF23水平过高所致。获得性FGF23相关型肾性低磷血症中最常见的类型为肿瘤性骨软化症(tumor-induced osteomalacia,TIO),系肿瘤分泌过量FGF23所致,此种类型的肿瘤大多为磷酸盐尿性间叶性肿瘤(phosphaturic mesenchymal tumours,PMTs)[18]。此外,药物亦可引起FGF23水平增高导致低磷血症,其中较为明确的是静脉麦芽糖铁的应用[16],而一些使用核苷酸类抗病毒药的患者中亦有报道FGF23水平增高,因此FGF23可能为该类抗病毒药导致低磷血症的原因之一[19-20]
遗传性FGF23相关型肾性低磷血症中最为常见的类型是PHEX基因突变导致的X连锁低磷血症(X-linked hypophosphatemia,XLH)。XLH是由于一种磷调节中性肽链内切酶功能异常所致,其发病率为3.9/100 000活产新生儿,患病率为1.7/100 000(儿童)~4.8/100 000(儿童和成人)不等[21]。2019年国内1项对261例XLH患者临床和遗传分析的回顾性研究报道了4个突变热点,并对基因型和表型之间的关联进行了探究,这是国内最大的XLH患者队列研究[22]
3. PTH相关型肾性低磷血症: 各种原因导致PTH增多可引起尿磷排泄增加,血磷降低。甲状旁腺腺瘤所致的原发性甲状旁腺功能亢进症(甲旁亢)和钙摄入缺乏引起的继发性甲旁亢是PTH相关型肾性低磷血症的两大主要病因,而治疗骨质疏松的药物或肿瘤靶向药则可通过引起低血钙导致继发性甲旁亢引起低血磷,此外特立帕肽(PTH重组活性片段)可直接升高PTH浓度引起低磷血症[16]。遗传性疾病亦可导致PTH增高,但往往多伴有其他内分泌器官肿瘤形成[23-24],如多发性内分泌腺瘤病,其临床症状复杂,且低血磷非主要表现,血磷通常维持在正常值下限,波动于0.97~1.13 mmol/L[25],故表3未列出遗传性PTH增多性疾病。
4. NPT相关型肾性低磷血症: NPT相关型肾性低磷血症系NPT异常致尿磷重吸收减少引起血磷降低。获得性NPT相关型肾性低磷血症多由药物引起,阿昔洛韦和雌激素可下调肾脏近端小管NPT的表达,导致尿磷排泄增加继而出现低磷血症[16]。遗传性NPT相关型肾性低磷血症因NPT突变或维持NPT正常表达的蛋白(Na+/H+交换调节因子1,NHERF1)异常所致,以高尿钙、高活性维生素D和肾结石为主要特点,具体信息详见表3
5. 非NPT相关型肾小管损伤导致的肾性低磷血症: 此种类型的肾性低磷血症为药物导致肾性失磷的主要原因,多为小管上皮细胞受损所致。引起此类肾性低磷血症的药物有很多,包括多种化疗药及靶向药、抗癫痫药、氨基糖苷类抗生素、抗结核药、核苷类抗病毒药、蛋白酶抑制剂、非甾体类抗炎药等[16,26]。一篇关于5例长期阿德福韦单药治疗导致肾性低磷软骨病的研究显示,这些患者除了低血磷、高尿磷外,同时伴有肾性失钾、尿钙排泄增加、尿液酸化功能异常、肾性糖尿、中低分子量蛋白尿等近端小管受损的临床特征,肾活检则主要表现为小管坏死、上皮细胞空泡样变性、轻度间质纤维化伴炎症细胞浸润[27]。此种类型相关的遗传性疾病包括Dent病、范可尼综合征等,详细信息见表3

三、 低磷血症的病因诊断

低磷血症的诊断依据血磷水平,但其病因鉴别十分复杂,需依赖详细的病史询问、临床表现、实验室检验及影像学检查方可初步判断,怀疑遗传性疾病需明确疾病类型时往往依赖于基因检测。
病史询问应注意询问家族史、饮食中是否存在维生素D及钙的缺乏、是否有充足的阳光照射及是否存在影响吸收的其他疾病[28],对于儿童还应询问喂养史。
临床表现存在异质性,与发病年龄相关,其最主要的临床表现为佝偻病或软骨病(osteomalacia)。虽然二者均为低血磷或低血钙所导致骨矿化异常,但佝偻病侧重于干骺端新生骨的矿化障碍,因此仅发生于处于生长发育阶段、生长板未闭合的儿童。而软骨病则为已形成的骨(旧骨)在重构过程中出现的矿化障碍,这一概念曾经主要被用来描述生长发育停止之后出现的骨矿化障碍,但实际上在儿童和成人中均可发病[28-31]。佝偻病的临床表现常自下肢负重时开始出现,主要为身高受限、骨骼畸形(手足镯征、肋串珠、内翻外翻畸形等)、骨折、牙釉质发育不全及牙周脓肿等。软骨病则主要表现为骨痛、鸡胸、脊柱弯曲、假性骨折等[28,32],其中骨痛为全身性(包括关节部位)或限于载重部位,易误诊为风湿病或关节炎[33]。此外由于低磷血症可导致细胞内ATP合成减少、供能不足,故可导致神经、肌肉、血液系统等方面的相关表现,如感觉异常、四肢轻瘫[10,34]、肌无力[35]、横纹肌溶解[36]、溶血性贫血[37]、白细胞功能异常[38]等。
实验室检验应包含血尿肌酐、同步血尿电解质、碱性磷酸酶(AKP)、25-(OH)-D3、1,25-(OH)2-D3、PTH、FGF23等指标的测定。目前FGF23的检测形式主要有两种:全段FGF23和FGF23羧基端,以全段FGF23检测为主。此外判断患者是否存在肾性失磷对于明确低磷血症的病因十分关键,最大肾小管磷重吸收率与肾小球滤过率之比值(TmP/GFR)是明确患者是否存在肾性失磷的主要指标[17,21,28],其受去脂体重影响小[18],可参照如下公式计算,参考范围[39]表4
TmP/GFR={TRP×血磷,        TRP0.860.3×TRP10.8×TRP×血磷,      TRP>0.86
TRP=1-尿××尿
表4 最大肾小管磷重吸收率与肾小球滤过率之比值(TmP/GFR)参考范围
年龄 性别 参考范围(mmol/L)
出生 男性和女性 1.43~3.43
3个月 男性和女性 1.48~3.30
6个月 男性和女性 1.15~2.60
2~15岁 男性和女性 1.15~2.44
25~35岁
男性 1.00~1.35
女性 0.96~1.44
45~55岁
男性 0.90~1.35
女性(绝经前) 0.88~1.42
65~75岁 男性和女性 0.80~1.35
影像学检查可为佝偻病和软骨病提供证据,佝偻病X线片的主要表现为腕关节或膝关节生长板增宽、干骺端杯状凹陷或边缘毛糙,其严重程度可依据Thacher佝偻病严重程度评分(rickets severity score,RSS)进行评定[40-41],而软骨病则可通过X线片的假骨折线(Looser's zone)[33]、放射性核素骨显像的多重摄取[42]辅以鉴别。若怀疑肿瘤性骨软化症(TIO),则需同时完善功能显像(如111铟标记的奥曲肽SPECT-CT)和解剖成像(如CT、MRI)检查等,以排查肿瘤[18]
Haffner等[21]提出低磷血症可依据调磷激素及尿磷排泄水平进行病因分类(图2)。当患者存在低磷血症时,PTH水平的升高主要指向同时合并低钙、甲状旁腺功能亢进的疾病,即维生素D相关型肾性低磷血症和PTH相关型肾性低磷血症。若PTH处于正常或偏低范围,则进一步计算患者TmP/GFR,若患者TmP/GFR不低,则说明患者不存在肾性失磷,其低磷的原因可能为磷摄入不足、肾外磷清除增多及体内磷的重分布所致。若患者TmP/GFR降低,再评估患者循环FGF23水平,如若FGF23升高则提示FGF23相关型肾性低磷血症,如若FGF23水平不高,则考虑NPT相关或小管功能受损导致的肾性低磷血症。如需进一步明确具体疾病类型,则需进行继发因素排查及基因检测。目前基因检测技术在遗传性肾性低磷血症的诊断中已有较为成熟的应用,基于二代测序技术开展的全外显子测序和全基因组测序已广泛用于临床,在罕见病和遗传性疾病致病基因鉴定方面取得巨大成功,同时也帮助克服了传统连锁分析方法的一些局限性[43]
图2 低磷血症病因诊断流程图
注:PTH:甲状旁腺素;FGF23:成纤维细胞生长因子23;NPT:钠磷共转运体

Full size|PPT slide

四、 肾性低磷血症的治疗原则及进展

肾性低磷血症的治疗最重要的一点是依据病因结合临床症状及相关检验检查有选择性进行治疗。获得性肾性低磷血症通常在消除获得性病因后即可改善,如停用导致低磷的药物,通过手术切除、放疗及消融等方式消除肿瘤,若停药或外科方法无法改善,则需进行内科治疗及饮食疗法。
常用的内科治疗药物有补磷制剂、维生素D制剂、钙剂和拟钙剂等,其中国内的口服补磷制剂主要为中性磷合剂,国外口服补磷制剂有溶液、胶囊、药片等多种形式,不同制剂含磷量差异很大,例如Joulies口服液中含磷元素30 mg/ml,而钾-磷中性药片每片包含磷元素250 mg[44],因此在补磷制剂选择上需依据不同制剂的实际含磷量决定每日用量。日常饮食中含磷量较高的食物有肉、奶、蛋、豆类及豆制品、坚果、种子、谷物和干货等。
维生素D相关型肾性低磷血症因其始动因素为维生素D不足或受体功能异常,常规不需补磷。维生素D摄入不足导致营养性佝偻病发生时,应在补钙的基础上根据年龄选择单次大剂量或90 d持续补充维生素D,较为推荐的是90 d疗法(详见表5),90 d后再次评估决定是否需要进一步治疗或是开始每日维持剂量治疗[28,30-31,45]。遗传性维生素D相关型肾性低磷血症需补充活性维生素D,VDDR2可尝试静脉补钙,其治疗剂量[28,46-52]表5,但VDDR2的秃顶患者对于维生素D或补钙治疗均无效[28]
表5 肾性低磷血症常规治疗总结
疾病名称(英文缩写) 维生素D
是否补充 推荐剂量
营养性佝偻病

90 d疗法(维生素D2/D3均可):年龄<12个月,2 000 IU/d;12个月≤年龄≤12岁,
3 000~6 000 IU/d;年龄>12岁,6 000 IU/d
维生素D依赖型
佝偻病1A(VDDR1A)

(骨化三醇或阿法骨化醇)
起始剂量:骨化三醇10~400 ng·kg-1·d-1或阿法骨化醇80~100 ng·kg-1·d-1
维生素D依赖型
佝偻病1B(VDDR1B)

(仅骨化三醇)
尚无
维生素D依赖型
佝偻病2A/2B(VDDR2A/2B)


尚无(有文献报道阿法骨化醇20~200 μg/d或骨化三醇17~20 μg/d)
FGF23相关型肾性低磷
血症——以XLH为例


起始剂量:骨化三醇20~30 ng·kg-1·d-1,每日1~2次;或阿法骨化醇30~50 ng·kg-1·d-1,每日1次
NPT相关型/肾小管损伤
导致的肾性低磷血症
疾病名称(英文缩写) 补磷制剂 补钙制剂
是否补充 推荐剂量 是否补充 推荐剂量
营养性佝偻病 ≥500 mg/d
维生素D依赖型
佝偻病1A(VDDR1A)
维生素D依赖型
佝偻病1B(VDDR1B)
维生素D依赖型
佝偻病2A/2B(VDDR2A/2B)
√(维生素D治疗
无效时可尝试使用)
400~1 400 mg·(m2)-1·d-1
静脉应用
FGF23相关型肾性低磷
血症——以XLH为例

起始剂量:20~60 mg·kg-1·d-1,一日4~6次
(高AKP水平)或一日3~4次(AKP正常)
NPT相关型/肾小管损伤
导致的肾性低磷血症

尚无
注:XLH:X连锁低磷血症;NPT:钠磷共转运体;AKP:碱性磷酸酶
非手术治疗的原发性甲旁亢可用西那卡塞降低血钙和PTH,二膦酸盐改善骨密度,若存在维生素D缺乏则需同时补充维生素D[25]
NPT相关型肾性低磷血症患者因其体内活性维生素D偏高同时尿钙增高,故仅需单纯补磷,补磷的目标是降低体内升高的活性维生素D的水平,从而减少小肠对钙的吸收,缓解高尿钙。肾小管功能受损导致的低磷需在补磷的基础上同时纠正其他电解质及酸碱平衡的紊乱。
FGF23相关型肾性低磷血症因其低血磷、低活性维生素D,故需同时补磷及活性维生素D以达到改善骨骼畸形及促进生长发育(儿童)或改善血磷水平(成人)的目标。以XLH儿童为例,补磷的起始剂量推荐为20~60 mg·kg-1·d-1,而骨化三醇的推荐补充剂量为20~30 ng·kg-1·d-1,需监测血磷、血钙、尿钙排泄率、AKP、PTH等调整用量[21,28,44,53]
长期的肾性低磷血症会引起一系列并发症,如继发性/三发性甲旁亢、XLH患者可能出现的高血压和左室肥厚[54]等。继发性甲旁亢可能与FGF23和磷酸盐制剂对甲状旁腺细胞长期刺激及低活性维生素D水平有关,因此需增加活性维生素D的补充剂量同时减少磷酸盐的补充。若治疗效果欠佳可考虑联合西那卡塞治疗,但西那卡塞并未获批用于该适应证,且可引起低钙血症和QT间期延长等不良反应,故使用时需密切监测。三发性高钙血症性甲旁亢患者应考虑甲状旁腺切除术治疗。对于XLH患者的高血压和左室肥厚,因研究中对其是否存在报道不一,故2019年由Haffner等[21]编写的XLH诊疗临床共识推荐对XLH患者至少每年监测一次血压,并在血压持续升高的情况下方可进行更为详细的心血管系统评估,而有关心血管并发症的治疗效果因缺少数据目前尚不可知。
2018年针对FGF23的完全人源化单克隆IgG1抗体Burosumab的上市为低磷血症性佝偻病的治疗带来新突破,该单抗目前获批用于治疗1岁及以上儿童和成人的XLH,其儿童的3期临床试验被证实可以改善佝偻病的严重程度、促进生长发育、提升活动能力及改善低磷性佝偻病相关血清学指标[55],成人3期临床试验则表明该单抗对于改善血磷水平、治愈骨折疗效显著[56]。欧洲药品管理局(EMA)和美国食品药品监督管理局(FDA)批准Burosumab用于儿童的起始治疗剂量分别为0.4 mg/kg体重和0.8 mg/kg体重,每2周皮下注射一次维持治疗,剂量可根据血磷水平按照每次0.4 mg/kg向上滴定,最大单次剂量为2 mg/kg体重(单次总剂量不超过90 mg)。成人XLH患者Burosumab起始治疗剂量为1 mg/kg体重,每4周皮下注射一次维持,剂量亦可递增,最大单次剂量为90 mg[21]。目前该单抗在TIO成人患者中的Ⅱ期开放标签临床试验正在进行中,Burosumab已于2020年6月获得FDA批准,用于治疗肿瘤不能定位或根治性切除的TIO引起的低磷血症,预期在不久的将来Burosumab将为TIO患者带来福音。

五、 总结

低磷血症在肾脏病中十分常见,肾脏是调控血磷的最主要器官,各种遗传性或获得性因素引起调磷激素水平异常、钠磷共转运体功能异常或肾小管损伤均可导致肾性失磷引起低磷血症,但目前临床工作者对于低磷血症病因的诊断及治疗缺乏系统性认知。低磷血症病因诊断依赖调磷激素及尿磷排泄水平的测定,治疗需因病制宜,而针对FGF23的单抗的出现则为特定类型低磷血症的治疗带来新选择、新希望。

References

[1]
Levin A, Bakris GL, Molitch M, et al. Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease[J]. Kidney Int, 2007, 71(1): 31-38. DOI: 10.1038/sj.ki.5002009.
Abnormalities of mineral metabolism occur early in chronic kidney disease. Quantification of the prevalence of these abnormalities has not been described using current assays nor in large unselected populations. This outpatient cohort cross-sectional study was conducted in 153 centers, (71% primary care practices). Blood for parathyroid hormone (PTH), vitamin D metabolites, creatinine, calcium (Ca), and phosphorus (P) were drawn between June and October 2004. Low 1,25-dihydroxyvitamin D (1,25 OH2 D3) was defined as <22 pg/ml. The 1814 patients had a mean age of 71.1 years old; 48% had diabetes mellitus (DM). Low 1,25 OH2 D3 was evident at all estimated glomerular filtration rate (eGFR) levels: 13% in those with eGFR >80 ml/min, >60% in those with eGFR <30 ml/min. High PTH (>65pm/dl) occurred in 12% with eGFR >80 ml/min. Serum Ca and P were normal until eGFR was <40 ml/min. Significant differences in the mean and median values of 1,25 OH2 D3, PTH, but not 25(OH)D3 levels, were seen across deciles of eGFR (P<0.001). Multivariate analysis revealed that DM, increased urinary albumin/creatinine ratio and lower eGFR predicted lower values of 1,25 OH2 D3. A high prevalence of mineral metabolite abnormalities occurs in a large unreferred US cohort. Low 1,25 OH2 D3 and elevated PTH are common at higher eGFR than previously described. As bone, cardiovascular disease, and mineral metabolite are correlated; further studies are necessary to determine the importance of these findings relative to outcomes.
[2]
Kidney Disease: Improving Global Outcomes KDIGO CKD-MBD Work Group. KDIGO clinical practice guideline for the diagnosis, evaluation, prevention, and treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD)[J]. Kidney Int Suppl, 2009, (113): S1-S130. DOI: 10.1038/ki.2009.188.
[3]
Kidney Disease: Improving Global Outcomes CKDMBDUWG. KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD)[J]. Kidney Int Suppl (2011), 2017, 7(1):1-59. DOI: 10.1016/j.kisu.2017.04.001.
[4]
国家肾脏疾病临床医学研究中心. 中国慢性肾脏病矿物质和骨异常诊治指南概要[J]. 肾脏病与透析肾移植杂志, 2019, 28(1): 52-57. DOI: 10.3969/j.issn.1006-298X.2019.01.012.
[5]
耿雪梅, 王一梅, 章晓燕, 等. 不同血清磷水平对综合性医院住院患者和慢性肾脏病患者病死率的影响[J]. 上海医学, 2017, 40(1): 14-20.
[6]
张庆, 蔡阳平, 刘红栓, 等. 早期低磷血症对重症患者预后的影响[J]. 中国继续医学教育, 2018, 10(35): 80-83. DOI: 10.3969/j.issn.1674-9308.2018.35.033.
[7]
Wagner CA, Rubio-Aliaga I, Biber J, et al. Genetic diseases of renal phosphate handling[J]. Nephrol Dial Transplant, 2014, 29 Suppl 4: iv45-iv54. DOI: 10.1093/ndt/gfu217.
[8]
Hernando N, Wagner CA. Mechanisms and regulation of intestinal phosphate absorption[J]. Compr Physiol, 2018, 8(3): 1065-1090. DOI: 10.1002/cphy.c170024.
States of hypo- and hyperphosphatemia have deleterious consequences including rickets/osteomalacia and renal/cardiovascular disease, respectively. Therefore, the maintenance of appropriate plasma levels of phosphate is an essential requirement for health. This control is executed by the collaborative action of intestine and kidney whose capacities to (re)absorb phosphate are regulated by a number of hormonal and metabolic factors, among them parathyroid hormone, fibroblast growth factor 23, 1,25(OH) vitamin D, and dietary phosphate. The molecular mechanisms responsible for the transepithelial transport of phosphate across enterocytes are only partially understood. Indeed, whereas renal reabsorption entirely relies on well-characterized active transport mechanisms of phosphate across the renal proximal epithelia, intestinal absorption proceeds via active and passive mechanisms, with the molecular identity of the passive component still unknown. The active absorption of phosphate depends mostly on the activity and expression of the sodium-dependent phosphate cotransporter NaPi-IIb (SLC34A2), which is highly regulated by many of the factors, mentioned earlier. Physiologically, the contribution of NaPi-IIb to the maintenance of phosphate balance appears to be mostly relevant during periods of low phosphate availability. Therefore, its role in individuals living in industrialized societies with high phosphate intake is probably less relevant. Importantly, small increases in plasma phosphate, even within normal range, associate with higher risk of cardiovascular disease. Therefore, therapeutic approaches to treat hyperphosphatemia, including dietary phosphate restriction and phosphate binders, aim at reducing intestinal absorption. Here we review the current state of research in the field. © 2017 American Physiological Society. Compr Physiol 8:1065-1090, 2018.Copyright © 2018 American Physiological Society. All rights reserved.
[9]
Biber J, Hernando N, Forster I. Phosphate transporters and their function[J]. Annu Rev Physiol, 2013, 75: 535-550. DOI: 10.1146/annurev-physiol-030212-183748.
Plasma phosphate concentration is maintained within a relatively narrow range by control of renal reabsorption of filtered inorganic phosphate (P(i)). P(i) reabsorption is a transcellular process that occurs along the proximal tubule. P(i) flux at the apical (luminal) brush border membrane represents the rate-limiting step and is mediated by three Na(+)-dependent P(i) cotransporters (members of the SLC34 and SLC20 families). The putative proteins responsible for basolateral P(i) flux have not been identified. The transport mechanism of the two kidney-specific SLC34 proteins (NaPi-IIa and NaPi-IIc) and of the ubiquitously expressed SLC20 protein (PiT-2) has been studied by heterologous expression to reveal important differences in kinetics, stoichiometry, and substrate specificity. Studies on the regulation of the abundance of the respective proteins highlight significant differences in the temporal responses to various hormonal and nonhormonal factors that can influence P(i) homeostasis. The phenotypes of mice deficient in NaPi-IIa and NaPi-IIc indicate that NaPi-IIa is responsible for most P(i) renal reabsorption. In contrast, in the human kidney, NaPi-IIc appears to have a relatively greater role. The physiological relevance of PiT-2 to P(i) reabsorption remains to be elucidated.
[10]
Christov M, Jüppner H. Phosphate homeostasis disorders[J]. Best Pract Res Clin Endocrinol Metab, 2018, 32(5): 685-706. DOI: 10.1016/j.beem.2018.06.004.
Our understanding of the regulation of phosphate balance has benefited tremendously from the molecular identification and characterization of genetic defects leading to a number of rare inherited or acquired disorders affecting phosphate homeostasis. The identification of the key phosphate-regulating hormone, fibroblast growth factor 23 (FGF23), as well as other molecules that control its production, such as the glycosyltransferase GALNT3, the endopeptidase PHEX, and the matrix protein DMP1, and molecules that function as downstream effectors of FGF23 such as the longevity factor Klotho and the phosphate transporters NPT2a and NPT2c, has permitted us to understand the complex interplay that exists between the kidneys, bone, parathyroid, and gut. Such insights from genetic disorders have allowed not only the design of potent targeted treatment of FGF23-dependent hypophosphatemic conditions, but also provide clinically relevant observations related to the dysregulation of mineral ion homeostasis in health and disease.Copyright © 2018. Published by Elsevier Ltd.
[11]
Wagner CA, Rubio-Aliaga I, Hernando N. Renal phosphate handling and inherited disorders of phosphate reabsorption: an update[J]. Pediatr Nephrol, 2019, 34(4): 549-559. DOI: 10.1007/s00467-017-3873-3.
Renal phosphate handling critically determines plasma phosphate and whole body phosphate levels. Filtered phosphate is mostly reabsorbed by Na-dependent phosphate transporters located in the brush border membrane of the proximal tubule: NaPi-IIa (SLC34A1), NaPi-IIc (SLC34A3), and Pit-2 (SLC20A2). Here we review new evidence for the role and relevance of these transporters in inherited disorders of renal phosphate handling. The importance of NaPi-IIa and NaPi-IIc for renal phosphate reabsorption and mineral homeostasis has been highlighted by the identification of mutations in these transporters in a subset of patients with infantile idiopathic hypercalcemia and patients with hereditary hypophosphatemic rickets with hypercalciuria. Both diseases are characterized by disturbed calcium homeostasis secondary to elevated 1,25-(OH) vitamin D as a consequence of hypophosphatemia. In vitro analysis of mutated NaPi-IIa or NaPi-IIc transporters suggests defective trafficking underlying disease in most cases. Monoallelic pathogenic mutations in both SLC34A1 and SLC34A3 appear to be very frequent in the general population and have been associated with kidney stones. Consistent with these findings, results from genome-wide association studies indicate that variants in SLC34A1 are associated with a higher risk to develop kidney stones and chronic kidney disease, but underlying mechanisms have not been addressed to date.
[12]
Bitzan M, Goodyer PR. Hypophosphatemic rickets[J]. Pediatr Clin North Am, 2019, 66(1): 179-207. DOI: 10.1016/j.pcl.2018.09.004.
[13]
Kliegman RM, Stanton B, St. Geme J, et al. Nelson's textbook of pediatrics[M]. 20th ed. Philadelphia: Elsevier, 2016.
[14]
Levi M, Gratton E, Forster IC, et al. Mechanisms of phosphate transport[J]. Nat Rev Nephrol, 2019, 15(8): 482-500. DOI: 10.1038/s41581-019-0159-y.
Over the past 25 years, successive cloning of SLC34A1, SLC34A2 and SLC34A3, which encode the sodium-dependent inorganic phosphate (P) cotransport proteins 2a-2c, has facilitated the identification of molecular mechanisms that underlie the regulation of renal and intestinal P transport. P and various hormones, including parathyroid hormone and phosphatonins, such as fibroblast growth factor 23, regulate the activity of these P transporters through transcriptional, translational and post-translational mechanisms involving interactions with PDZ domain-containing proteins, lipid microdomains and acute trafficking of the transporters via endocytosis and exocytosis. In humans and rodents, mutations in any of the three transporters lead to dysregulation of epithelial P transport with effects on serum P levels and can cause cardiovascular and musculoskeletal damage, illustrating the importance of these transporters in the maintenance of local and systemic P homeostasis. Functional and structural studies have provided insights into the mechanism by which these proteins transport P, whereas in vivo and ex vivo cell culture studies have identified several small molecules that can modify their transport function. These small molecules represent potential new drugs to help maintain P homeostasis in patients with chronic kidney disease - a condition that is associated with hyperphosphataemia and severe cardiovascular and skeletal consequences.
[15]
Khundmiri SJ, Murray RD, Lederer E. PTH and vitamin D[J]. Compr Physiol, 2016, 6(2): 561-601. DOI: 10.1002/cphy.c140071.
PTH and Vitamin D are two major regulators of mineral metabolism. They play critical roles in the maintenance of calcium and phosphate homeostasis as well as the development and maintenance of bone health. PTH and Vitamin D form a tightly controlled feedback cycle, PTH being a major stimulator of vitamin D synthesis in the kidney while vitamin D exerts negative feedback on PTH secretion. The major function of PTH and major physiologic regulator is circulating ionized calcium. The effects of PTH on gut, kidney, and bone serve to maintain serum calcium within a tight range. PTH has a reciprocal effect on phosphate metabolism. In contrast, vitamin D has a stimulatory effect on both calcium and phosphate homeostasis, playing a key role in providing adequate mineral for normal bone formation. Both hormones act in concert with the more recently discovered FGF23 and klotho, hormones involved predominantly in phosphate metabolism, which also participate in this closely knit feedback circuit. Of great interest are recent studies demonstrating effects of both PTH and vitamin D on the cardiovascular system. Hyperparathyroidism and vitamin D deficiency have been implicated in a variety of cardiovascular disorders including hypertension, atherosclerosis, vascular calcification, and kidney failure. Both hormones have direct effects on the endothelium, heart, and other vascular structures. How these effects of PTH and vitamin D interface with the regulation of bone formation are the subject of intense investigation.Copyright © 2016 John Wiley & Sons, Inc.
[16]
Megapanou E, Florentin M, Milionis H, et al. Drug-induced hypophosphatemia: current insights[J]. Drug Saf, 2020, 43(3): 197-210. DOI: 10.1007/s40264-019-00888-1.
[17]
Acar S, Demir K, Shi Y. Genetic causes of rickets[J]. J Clin Res Pediatr Endocrinol, 2017, 9(Suppl 2): 88-105. DOI: 10.4274/jcrpe.2017.S008.
Rickets is a metabolic bone disease that develops as a result of inadequate mineralization of growing bone due to disruption of calcium, phosphorus and/or vitamin D metabolism. Nutritional rickets remains a significant child health problem in developing countries. In addition, several rare genetic causes of rickets have also been described, which can be divided into two groups. The first group consists of genetic disorders of vitamin D biosynthesis and action, such as vitamin D-dependent rickets type 1A (VDDR1A), vitamin D-dependent rickets type 1B (VDDR1B), vitamin D-dependent rickets type 2A (VDDR2A), and vitamin D-dependent rickets type 2B (VDDR2B). The second group involves genetic disorders of excessive renal phosphate loss (hereditary hypophosphatemic rickets) due to impairment in renal tubular phosphate reabsorption as a result of FGF23-related or FGF23-independent causes. In this review, we focus on clinical, laboratory and genetic characteristics of various types of hereditary rickets as well as differential diagnosis and treatment approaches.
[18]
Minisola S, Peacock M, Fukumoto S, et al. Tumour-induced osteomalacia[J]. Nat Rev Dis Primers, 2017, 3: 17044. DOI: 10.1038/nrdp.2017.44.
Tumour-induced osteomalacia (TIO), also known as oncogenic osteomalacia, is a rare paraneoplastic disorder caused by tumours that secrete fibroblast growth factor 23 (FGF23). Owing to the role of FGF23 in renal phosphate handling and vitamin D synthesis, TIO is characterized by decreased renal tubular reabsorption of phosphate, by hypophosphataemia and by low levels of active vitamin D. Chronic hypophosphataemia ultimately results in osteomalacia (that is, inadequate bone mineralization). The diagnosis of TIO is usually suspected when serum phosphate levels are chronically low in the setting of bone pain, fragility fractures and muscle weakness. Locating the offending tumour can be very difficult, as the tumour is often very small and can be anywhere in the body. Surgical removal of the tumour is the only definitive treatment. When the tumour cannot be located or when complete resection is not possible, medical treatment with phosphate salts or active vitamin D is necessary. One of the most promising emerging treatments for unresectable tumours that cause TIO is the anti-FGF23 monoclonal antibody KRN23. The recent identification of a fusion of fibronectin and fibroblast growth factor receptor 1 (FGFR1) as a molecular driver in some tumours not only sheds light on the pathophysiology of TIO but also opens the door to a better understanding of the transcription, translocation, post-translational modification and secretion of FGF23, as well as suggesting approaches to targeted therapy. Further study will reveal if the FGFR1 pathway is also involved in tumours that do not harbour the translocation.
[19]
Saeedi R, Jiang SY, Holmes DT, et al. Fibroblast growth factor 23 is elevated in tenofovir-related hypophosphatemia[J]. Calcif Tissue Int, 2014, 94(6): 665-668. DOI: 10.1007/s00223-014-9854-7.
[20]
Cheng CY, Chang SY, Lin MH, et al. Tenofovir disoproxil fumarate-associated hypophosphatemia as determined by fractional excretion of filtered phosphate in HIV-infected patients[J]. J Infect Chemother, 2016, 22(11): 744-747. DOI: 10.1016/j.jiac.2016.08.008.
[21]
Haffner D, Emma F, Eastwood DM, et al. Clinical practice recommendations for the diagnosis and management of X-linked hypophosphataemia[J]. Nat Rev Nephrol, 2019, 15(7): 435-455. DOI: 10.1038/s41581-019-0152-5.
X-linked hypophosphataemia (XLH) is the most common cause of inherited phosphate wasting and is associated with severe complications such as rickets, lower limb deformities, pain, poor mineralization of the teeth and disproportionate short stature in children as well as hyperparathyroidism, osteomalacia, enthesopathies, osteoarthritis and pseudofractures in adults. The characteristics and severity of XLH vary between patients. Because of its rarity, the diagnosis and specific treatment of XLH are frequently delayed, which has a detrimental effect on patient outcomes. In this Evidence-Based Guideline, we recommend that the diagnosis of XLH is based on signs of rickets and/or osteomalacia in association with hypophosphataemia and renal phosphate wasting in the absence of vitamin D or calcium deficiency. Whenever possible, the diagnosis should be confirmed by molecular genetic analysis or measurement of levels of fibroblast growth factor 23 (FGF23) before treatment. Owing to the multisystemic nature of the disease, patients should be seen regularly by multidisciplinary teams organized by a metabolic bone disease expert. In this article, we summarize the current evidence and provide recommendations on features of the disease, including new treatment modalities, to improve knowledge and provide guidance for diagnosis and multidisciplinary care.
[22]
Zhang C, Zhao Z, Sun Y, et al. Clinical and genetic analysis in a large Chinese cohort of patients with X-linked hypophosphatemia[J]. Bone, 2019, 121: 212-220. DOI: 10.1016/j.bone.2019.01.021.
X-linked Hypophosphatemia (XLH) is caused by loss of function mutations in the PHEX gene. Given the recent availability of a new therapy for XLH, a retrospective analysis of the most recent 261 Chinese patients with XLH evaluated at Peking Union Medical College Hospital was conducted. Clinical, biochemical, radiographic studies, as well as genetic analyses, including Sanger sequencing for point mutations and Multiplex Ligation-dependent Probe Amplification (MLPA) to detect large deletions/duplications were employed. Based on the structure of Neprilysin (NEP), a member of M13 family that includes PHEX, a three-dimensional (3D) model of PHEX was constructed, missense and nonsense mutations were positioned on the predicted structure to visualize relative positions of these two types of variants. Sex differences and genotype-phenotype correlations were also undertaken. Genetic analyses identified 166 PHEX mutations in 261 XLH patients. One hundred and eleven of the 166 mutations were unreported. Four mutational 'hot-spots' were identified in this cohort (P534L, G579R, R747X, c.1645+1 G>A). Missense mutations, but not nonsense mutations, clustered in the two putative lobes of the PHEX protein, suggesting these are functionally important regions of the molecule. Circulating levels of intact FGF23 were significantly elevated (median level 101.9 pg/mL; reference range 16.1-42.2 pg/mL). No significant sex differences, as well as no phenotypic differences were identified between patients with putative truncating and non-truncating PHEX mutations. However, patients with N-terminal PHEX mutations had an earlier age of onset of disease (P = 0.015) and higher iFGF23 levels (P = 0.045) as compared to those with C-terminal mutations. These data provide a comprehensive characterization of the largest cohort of patients with XLH reported to date from China, which will help in evaluating the applicability of emerging therapies for this disease in this ethnic group.Copyright © 2019 Elsevier Inc. All rights reserved.
[23]
DeLellis RA, Mangray S. Heritable forms of primary hyperparathyroidism: a current perspective[J]. Histopathology, 2018, 72(1): 117-132. DOI: 10.1111/his.13306.
Primary hyperparathyroidism (PHPT) is one of the most common of all endocrine disorders encountered by the practising histopathologist. The vast majority of lesions are sporadic in nature, approximately 85% of which are parathyroid adenomas, while hyperplasia and carcinoma account for 10-15% and fewer than 1%, of cases, respectively. Heritable forms of PHPT are much less common and present challenges both to clinicians and pathologists, particularly when they are the presenting feature of an endocrine syndrome. In such instances, pathologists play a key role in alerting physicians to the possibility of an underlying heritable endocrine syndrome and the potential for extra-endocrine manifestations. Therefore, a working knowledge of these disorders is essential for providing guidance to treating physicians. The aim of this update is to review the clinicopathological features, genetic bases and current management for patients with PHPT associated with multiple endocrine neoplasia (MEN) types 1, 2A and 4 and hyperparathyroidism-jaw tumour (HPT-JT) syndrome in the context of the 2017 World Health Organization (WHO) Classification of Tumours of the Endocrine Organs. Additionally, familial isolated hyperparathyroidism, familial hypocalciuric hypercalcaemia and neonatal severe hyperparathyroidism are discussed.© 2017 John Wiley & Sons Ltd.
[24]
Marx SJ. Hyperparathyroid and hypoparathyroid disorders[J]. N Engl J Med, 2000, 343(25): 1863-1875. DOI: 10.1056/NEJM200012213432508.
[25]
Bilezikian JP, Bandeira L, Khan A, et al. Hyperparathyroidism[J]. Lancet, 2018, 391(10116): 168-178. DOI: 10.1016/S0140-6736(17)31430-7.
Primary hyperparathyroidism is a common endocrine disorder of calcium metabolism characterised by hypercalcaemia and elevated or inappropriately normal concentrations of parathyroid hormone. Almost always, primary hyperparathyroidism is due to a benign overgrowth of parathyroid tissue either as a single gland (80% of cases) or as a multiple gland disorder (15-20% of cases). Primary hyperparathyroidism is generally discovered when asymptomatic but the disease always has the potential to become symptomatic, resulting in bone loss and kidney stones. In countries where biochemical screening tests are not common, symptomatic primary hyperparathyroidism tends to predominate. Another variant of primary hyperparathyroidism has been described in which the serum calcium concentration is within normal range but parathyroid hormone is elevated in the absence of any obvious cause. Primary hyperparathyroidism can be cured by removal of the parathyroid gland or glands but identification of patients who are best advised to have surgery requires consideration of the guidelines that are regularly updated. Recommendations for patients who do not undergo parathyroid surgery include monitoring of serum calcium concentrations and bone density.Copyright © 2018 Elsevier Ltd. All rights reserved.
[26]
陈楠. 警惕生物制剂相关的肾脏损害[J]. 肾脏病与透析肾移植杂志, 2018, 27(6): 549-550. DOI: 10.3969/j.issn.1006-298X.2018.06.010.
[27]
Xu Y, Shen P, Pan X, et al. Nephrogenic hypophosphatemic osteomalacia during adefovir monotherapy for chronic hepatitis B monoinfection[J]. Clin Kidney J, 2013, 6(4): 379-383. DOI: 10.1093/ckj/sft069.
In this paper, we explore nephrogenic hypophosphatemic osteomalacia associated with low-dose adefovir dipivoxil (ADV) therapy.Five patients who were treated with ADV for >2 years were included in this study. The metabolic index of phosphate and calcium, renal tubular function, renal function and pathological changes of the patients were investigated.Two male and three female patients were studied. All of the patients presented with a reduced serum phosphate level (0.38-0.60 mmol/L) accompanied with hyperphosphaturia at 10.9-23.8 mmol/24 h. The serum potassium level was also reduced or at lower range (2.56-3.54 mmol/L), but the 24-h urinary potassium was relatively increased. Urinalysis also demonstrated increased excretion of glucose in four patients. Urine protein electrophoresis showed low-to-moderate molecular weight protein. Three patients manifested urine acidification function impairment. Four patients had accompanying renal insufficiency. Three patients had difficulty walking and presented with a reduction in height (2.5-14 cm). Renal biopsy revealed that most of the glomeruli were normal accompanied by mild interstitial fibrosis with inflammatory cell infiltration. ADV treatment was subsequently ceased. Patients were treated with regular phosphate supplementation, citrate acid potassium and calcium bicarbonate. After 6-month treatment, the bone pain was significantly alleviated. Serum creatinine of one patient returned to normal levels and two patients who had difficulty walking were able to walk independently.The current study showed long-term and low-dose ADV treatment in a Chinese population may lead to proximal tubular impairment, metabolic acidosis, hypophosphatemia, hypokalemia, metabolic bone disease, renal osteopathia and renal functional damage.
[28]
Carpenter TO, Shaw NJ, Portale AA, et al. Rickets[J]. Nat Rev Dis Primers, 2017, 3: 17101. DOI: 10.1038/nrdp.2017.101.
Rickets is a bone disease associated with abnormal serum calcium and phosphate levels. The clinical presentation is heterogeneous and depends on the age of onset and pathogenesis but includes bowing deformities of the legs, short stature and widening of joints. The disorder can be caused by nutritional deficiencies or genetic defects. Mutations in genes encoding proteins involved in vitamin D metabolism or action, fibroblast growth factor 23 (FGF23) production or degradation, renal phosphate handling or bone mineralization have been identified. The prevalence of nutritional rickets has substantially declined compared with the prevalence 200 years ago, but the condition has been re-emerging even in some well-resourced countries; prematurely born infants or breastfed infants who have dark skin types are particularly at risk. Diagnosis is usually established by medical history, physical examination, biochemical tests and radiography. Prevention is possible only for nutritional rickets and includes supplementation or food fortification with calcium and vitamin D either alone or in combination with sunlight exposure. Treatment of typical nutritional rickets includes calcium and/or vitamin D supplementation, although instances infrequently occur in which phosphate repletion may be necessary. Management of heritable types of rickets associated with defects in vitamin D metabolism or activation involves the administration of vitamin D metabolites. Oral phosphate supplementation is usually indicated for FGF23-independent phosphopenic rickets, whereas the conventional treatment of FGF23-dependent types of rickets includes a combination of phosphate and activated vitamin D; an anti-FGF23 antibody has shown promising results and is under further study.
[29]
Tiosano D, Hochberg Z. Hypophosphatemia: the common denominator of all rickets[J]. J Bone Miner Metab, 2009, 27(4): 392-401. DOI: 10.1007/s00774-009-0079-1.
Rickets is a disease of the hypertrophic chondrocytes in the growth plate and is caused by hypophosphatemia-a derived defect in terminal chondrocyte apoptosis. This highlights the critical role of phosphorous in cartilage and bone metabolism. This review shows the role of phosphorous metabolism, transport and function in maintaining phosphorous supply to the growth plate, bone osteoblast and the kidney. Given that phosphorous is the common denominator of all rickets, this review proposes a new classification for the differential diagnosis of rickets, which is based on the mechanisms leading to hypophosphatemia-high PTH activity, high FGF23 activity or renal phosphaturia.
[30]
Uday S, Högler W. Nutritional rickets and osteomalacia in the twenty-first century: revised concepts, public health, and prevention strategies[J]. Curr Osteoporos Rep, 2017, 15(4): 293-302. DOI: 10.1007/s11914-017-0383-y.
Nutritional rickets and osteomalacia are common in dark-skinned and migrant populations. Their global incidence is rising due to changing population demographics, failing prevention policies and missing implementation strategies. The calcium deprivation spectrum has hypocalcaemic (seizures, tetany and dilated cardiomyopathy) and late hypophosphataemic (rickets, osteomalacia and muscle weakness) complications. This article reviews sustainable prevention strategies and identifies areas for future research.The global rickets consensus recognises the equal contribution of vitamin D and dietary calcium in the causation of calcium deprivation and provides a three stage categorisation for sufficiency, insufficiency and deficiency. For rickets prevention, 400 IU daily is recommended for all infants from birth and 600 IU in pregnancy, alongside monitoring in antenatal and child health surveillance programmes. High-risk populations require lifelong supplementation and food fortification with vitamin D or calcium. Future research should identify the true prevalence of rickets and osteomalacia, their role in bone fragility and infant mortality, and best screening and public health prevention tools.
[31]
Munns CF, Shaw N, Kiely M, et al. Global consensus recommendations on prevention and management of nutritional rickets[J]. J Clin Endocrinol Metab, 2016, 101(2): 394-415. DOI: 10.1210/jc.2015-2175.
Vitamin D and calcium deficiencies are common worldwide, causing nutritional rickets and osteomalacia, which have a major impact on health, growth, and development of infants, children, and adolescents; the consequences can be lethal or can last into adulthood. The goals of this evidence-based consensus document are to provide health care professionals with guidance for prevention, diagnosis, and management of nutritional rickets and to provide policy makers with a framework to work toward its eradication.A systematic literature search examining the definition, diagnosis, treatment, and prevention of nutritional rickets in children was conducted. Evidence-based recommendations were developed using the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) system that describe the strength of the recommendation and the quality of supporting evidence.Thirty-three nominated experts in pediatric endocrinology, pediatrics, nutrition, epidemiology, public health, and health economics evaluated the evidence on specific questions within five working groups. The consensus group, representing 11 international scientific organizations, participated in a multiday conference in May 2014 to reach a global evidence-based consensus.This consensus document defines nutritional rickets and its diagnostic criteria and describes the clinical management of rickets and osteomalacia. Risk factors, particularly in mothers and infants, are ranked, and specific prevention recommendations including food fortification and supplementation are offered for both the clinical and public health contexts.Rickets, osteomalacia, and vitamin D and calcium deficiencies are preventable global public health problems in infants, children, and adolescents. Implementation of international rickets prevention programs, including supplementation and food fortification, is urgently required.
[32]
Reginato AJ, Coquia JA. Musculoskeletal manifestations of osteomalacia and rickets[J]. Best Pract Res Clin Rheumatol, 2003, 17(6): 1063-1080. DOI: 10.1016/j.berh.2003.09.004.
[33]
董德长. 30例软骨病临床分析[J]. 上海医学, 1981, (4): 21-26+61+63.
[34]
Michell AW, Burn DJ, Reading PJ. Central pontine myelinolysis temporally related to hypophosphataemia[J]. J Neurol Neurosurg Psychiatry, 2003, 74(6): 820. DOI: 10.1136/jnnp.74.6.820.
[35]
Veilleux LN, Cheung M, Ben Amor M, et al. Abnormalities in muscle density and muscle function in hypophosphatemic rickets[J]. J Clin Endocrinol Metab, 2012, 97(8): E1492-E1498. DOI: 10.1210/jc.2012-1336.
[36]
Singhal PC, Kumar A, Desroches L, et al. Prevalence and predictors of rhabdomyolysis in patients with hypophosphatemia[J]. Am J Med, 1992, 92(5): 458-464. DOI: 10.1016/0002-9343(92)90740-3.
We undertook this study to determine the prevalence and predictors of rhabdomyolysis in the hypophosphatemic state.To identify patients with hypophosphatemia, we reviewed medical admissions for the period of January through December 1989. The hypophosphatemic state was considered whenever the serum phosphate was less than or equal to 2.0 mg/dL. Rhabdomyolysis secondary to hypophosphatemia was defined when serum creatine kinase levels were greater than or equal to 224 IU/L; it occurred within 72 hours of the hypophosphatemic episode; and it subsequently normalized. Patients who had any other independent etiology for rhabdomyolysis were excluded. Clinical and biochemical characteristics of patients with rhabdomyolysis (Group I) and patients without rhabdomyolysis (Group II) were compared. Variables that predicted rhabdomyolysis in hypophosphatemia were identified by stepwise logistic regression using a backward elimination procedure.One hundred twenty-nine patients were found to have hypophosphatemia. Forty-six (Group I) of 129 patients (36%) showed biochemical evidence of rhabdomyolysis. There was no difference in serum phosphate and potassium concentrations between Group I and Group II patients. Patients in Group I showed higher values for serum osmolality (p less than 0.05), serum glutamic oxaloacetic transaminase (p less than 0.001), chloride (p less than 0.01), and blood urea nitrogen (less than 0.05) when compared with Group II patients. When biochemical profiles of patients with rhabdomyolysis were evaluated on the day of their peak creatine kinase level, only 16 patients were hypophosphatemic, and the majority of patients showed a transient increase in serum phosphate levels because of ongoing muscle cell injury. Of 17 potential predictors, six variables emerged including sodium, chloride, glucose, blood urea nitrogen, uric acid, and osmolality. These variables provided high sensitivity (0.88) as well as moderate specificity (0.79) for predicting the occurrence of rhabdomyolysis in hypophosphatemia.We conclude that rhabdomyolysis commonly occurs in the hypophosphatemic state and that at times severe hypophosphatemia as an etiology may be masked because of ongoing rhabdomyolysis. Serum sodium, chloride, glucose, blood urea nitrogen, uric acid, and osmolality have a predictive role for the occurrence of rhabdomyolysis in the hypophosphatemic state that shows a high specificity and a moderate sensitivity.
[37]
Melvin JD, Watts RG. Severe hypophosphatemia: a rare cause of intravascular hemolysis[J]. Am J Hematol, 2002, 69(3): 223-224. DOI: 10.1002/ajh.10071.
A 3-year-old child presented with severe hyperphosphatemia (phosphate 45 mg/dL) secondary to chronic enema use. Following aggressive correction of the hyperphosphatemia, hypophosphatemia ensued (phosphate 1.7 mg/dL). Concurrently, the patient developed severe intravascular hemolysis and RBC morphologic defects. The hemolysis and morphologic defects corrected with return to normal serum phosphate levels. Severe hypophosphatemia is a rare cause of intravascular hemolysis.
[38]
Craddock PR, Yawata Y, VanSanten L, et al. Acquired phagocyte dysfunction. A complication of the hypophosphatemia of parenteral hyperalimentation[J]. N Engl J Med, 1974, 290(25): 1403-1407. DOI: 10.1056/NEJM197406202902504.
[39]
Payne RB. Renal tubular reabsorption of phosphate (TmP/GFR): indications and interpretation[J]. Ann Clin Biochem, 1998, 35(Pt 2): 201-206. DOI: 10.1177/000456329803500203.
[40]
Thacher TD, Pettifor JM, Tebben PJ, et al. Rickets severity predicts clinical outcomes in children with X-linked hypophosphatemia: utility of the radiographic rickets severity Score[J]. Bone, 2019, 122: 76-81. DOI: 10.1016/j.bone.2019.02.010.
The Rickets Severity Score (RSS) was used to evaluate X-linked hypophosphatemic rickets (XLH), a genetic disorder mediated by increased circulating FGF23. The reliability of the RSS was assessed using data from a randomized, phase 2 clinical trial that evaluated the effects of burosumab, a fully human anti-FGF23 monoclonal antibody, in 52 children with XLH ages 5 to 12 years. Bilateral knee and wrist radiographs were obtained at baseline, week 40, and week 64. We evaluated the relationships of the RSS to the Radiographic Global Impression of Change (RGI-C), serum alkaline phosphatase (ALP), height Z-score, 6-minute walk test (6MWT) percent predicted, and the Pediatric Orthopedic Society of North America Pediatric Outcomes Data Collection Instrument (POSNA-PODCI). The RSS showed moderate-to-substantial inter-rater reliability (weighted kappa, 0.45-0.65; Pearson correlation coefficient (r), 0.83-0.89) and substantial intra-rater reliability (weighted Kappa, 0.66; r = 0.91). Baseline RSS correlated with serum ALP (r = 0.47). Baseline RSS identified two subgroups (higher [RSS ≥1.5] and lower RSS [RSS <1.5]) that discriminated between subjects with greater and lesser rachitic disease. Higher RSS was associated with more severe clinical features, including impaired growth (Z-score, -2.12 vs -1.44) and walking ability (6MWT percent predicted, 77% vs 86%), more severe self-reported pain (29.9 [more severe] vs 45.3 [less severe]) and less physical function (29.6 [more severe] vs 40.9 [less severe]). During burosumab treatment, greater reductions in RSS corresponded to higher RGI-C global scores (r = -0.65). Improvements in RSS correlated with decreased serum ALP (r = 0.47). These results show the reliability of the RSS in XLH, and demonstrate that higher RSS values are associated with greater biochemical, clinical, and functional impairments in children with XLH.Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.
[41]
Thacher TD, Fischer PR, Pettifor JM, et al. Radiographic scoring method for the assessment of the severity of nutritional rickets[J]. J Trop Pediatr, 2000, 46(3): 132-139. DOI: 10.1093/tropej/46.3.132.
[42]
Fukumoto S, Ozono K, Michigami T, et al. Pathogenesis and diagnostic criteria for rickets and osteomalacia--proposal by an expert panel supported by the Ministry of Health, Labour and Welfare, Japan, the Japanese Society for Bone and Mineral Research, and the Japan Endocrine Society[J]. J Bone Miner Metab, 2015, 33(5): 467-473. DOI: 10.1007/s00774-015-0698-7.
Rickets and osteomalacia are diseases characterized by impaired mineralization of bone matrix. Recent investigations have revealed that the causes of rickets and osteomalacia are quite variable. Although these diseases can severely impair the quality of life of affected patients, rickets and osteomalacia can be completely cured or at least respond to treatment when properly diagnosed and treated according to the specific causes. On the other hand, there are no standard criteria to diagnose rickets or osteomalacia nationally and internationally. Therefore, we summarize the definition and pathogenesis of rickets and osteomalacia, and propose diagnostic criteria and a flowchart for the differential diagnosis of various causes of these diseases. We hope that these criteria and the flowchart are clinically useful for the proper diagnosis and management of these diseases.
[43]
张强. 全外显子测序技术在罕见及复杂疾病中的应用[J]. 临床医药文献电子杂志, 2019, 6(79): 192.
[44]
Carpenter TO, Imel EA, Holm IA, et al. A clinician's guide to X-linked hypophosphatemia[J]. J Bone Miner Res, 2011, 26(7): 1381-1388. DOI: 10.1002/jbmr.340.
X-linked hypophosphatemia (XLH) is the prototypic disorder of renal phosphate wasting, and the most common form of heritable rickets. Physicians, patients, and support groups have all expressed concerns about the dearth of information about this disease and the lack of treatment guidelines, which frequently lead to missed diagnoses or mismanagement. This perspective addresses the recommendation by conferees for the dissemination of concise and accessible treatment guidelines for clinicians arising from the Advances in Rare Bone Diseases Scientific Conference held at the NIH in October 2008. We briefly review the clinical and pathophysiologic features of the disorder and offer this guide in response to the conference recommendation, based on our collective accumulated experience in the management of this complex disorder.Copyright © 2011 American Society for Bone and Mineral Research.
[45]
Creo AL, Thacher TD, Pettifor JM, et al. Nutritional rickets around the world: an update[J]. Paediatr Int Child Health, 2017, 37(2): 84-98. DOI: 10.1080/20469047.2016.1248170.
[46]
Reade TM, Scriver CR, Glorieux FH, et al. Response to crystalline 1alpha-hydroxyvitamin D3 in vitamin D dependency[J]. Pediatr Res, 1975, 9(7): 593-599. DOI: 10.1203/00006450-197507000-00008.
The therapeutic response to chemically synthesized 1alpha-hydroxycholecalciferol (1alpha-OH-D3) was studied in three patients with autosomal recessive vitamin D dependency (ARVDD). The daily maintenance dose for vitamin D2, to prevent signs of vitamin D deficiency in these patients, was 40-54.4 mug/kg, or about 100 times normal (Table 1). Withdrawal of maintenance therapy with vitamin D2 resulted in the ultimate reappearance of the vitamin D depletion syndrome in patients 1 and 2 (Figs. 1 and 2). The third patient presented with the deficiency syndrome despite adequate vitamin D nutrition and was recognized to have ARVDD. Treatment with 1alpha-OH-D3 by mouth in all three patients at dose levels of 1-3 mug/24 hr (80-100 ng/kg) corrected hypocalcemia and suppressed parathyroid hormone-dependent renal loss of amino acids (Figs. 1, 2, and 4). Rickets healed in 7-9 weeks on 1alpha-OH-D3 alone (Fig. 3). The therapeutic response was rapid. It was usually seen first in the rise of serum calcium (Figs. 5 and 6). Withdrawal of 1alpha-OH-D3 was followed first by a fall of serum phosphorus, then by a fall in serum calcium; the latter occurred within about 2 weeks of withdrawal. Because the synthesis of 1alpha-OH-D3 is simpler than for 1alpha,25-dihydroxycholecalciferol and because the former is an effective therapeutic analog of vitamin D hormone, we believe these studies in ARVDD reveal 1alpha-OH-D3 to be the agent of choice for treatment of this and analogous diseases.
[47]
Marx SJ, Spiegel AM, Brown EM, et al. A familial syndrome of decrease in sensitivity to 1,25-dihydroxyvitamin D[J]. J Clin Endocrinol Metab, 1978, 47(6): 1303-1310. DOI: 10.1210/jcem-47-6-1303.
[48]
Balsan S, Garabédian M, Larchet M, et al. Long-term nocturnal calcium infusions can cure rickets and promote normal mineralization in hereditary resistance to 1,25-dihydroxyvitamin D[J]. J Clin Invest, 1986, 77(5): 1661-1667. DOI: 10.1172/JCI112483.
We report the beneficial effects of calcium infusions in a child with hereditary resistance to 1,25(OH)2D and alopecia. This patient after transient responsiveness to vitamin D derivatives became unresponsive to all therapy despite serum 1,25(OH)2D concentrations maintained at levels approximately 100-fold normal. A 7-mo trial with calcium infusions led to correction of biochemical abnormalities and healing of rickets. Bone biopsies (n = 3) showed a normal mineralization and the disappearance of the osteomalacia. Cultures of bone-derived cells demonstrated a lack of activation of 25-hydroxyvitamin D 24-hydroxylase and osteocalcin synthesis by 1,25(OH)2D3 (10(-9) and 10(-6) M). These results demonstrate that even in the absence of a normal 1,25(OH)2D3 receptor-effector system in bone cells, normal mineralization can be achieved in humans if adequate serum calcium and phosphorus concentrations are maintained; and calcium infusions may be an efficient alternative for the management of patients with this condition who are unresponsive to large doses of vitamin D derivatives.
[49]
Hochberg Z, Tiosano D, Even L. Calcium therapy for calcitriol-resistant rickets[J]. J Pediatr, 1992, 121(5 Pt 1):803-808. DOI: 10.1016/s0022-3476(05)81919-5.
[50]
Yagi H, Ozono K, Miyake H, et al. A new point mutation in the deoxyribonucleic acid-binding domain of the vitamin D receptor in a kindred with hereditary 1,25-dihydroxyvitamin D-resistant rickets[J]. J Clin Endocrinol Metab, 1993, 76(2): 509-512. DOI: 10.1210/jcem.76.2.8381803.
[51]
Ma NS, Malloy PJ, Pitukcheewanont P, et al. Hereditary vitamin D resistant rickets: identification of a novel splice site mutation in the vitamin D receptor gene and successful treatment with oral calcium therapy[J]. Bone, 2009, 45(4): 743-746. DOI: 10.1016/j.bone.2009.06.003.
To study the vitamin D receptor (VDR) gene in a young girl with severe rickets and clinical features of hereditary vitamin D resistant rickets, including hypocalcemia, hypophosphatemia, partial alopecia, and elevated serum levels of 1,25-dihydroxyvitamin D.We amplified and sequenced DNA samples from blood from the patient, her mother, and the patient's two siblings. We also amplified and sequenced the VDR cDNA from RNA isolated from the patient's blood.DNA sequence analyses of the VDR gene showed that the patient was homozygous for a novel guanine to thymine substitution in the 5'-splice site in the exon 8-intron J junction. Analysis of the VDR cDNA using reverse transcriptase-polymerase chain reaction showed that exons 7 and 9 were fused, and that exon 8 was skipped. The mother was heterozygous for the mutation and the two siblings were unaffected.A novel splice site mutation was identified in the VDR gene that caused exon 8 to be skipped. The mutation deleted amino acids 303-341 in the VDR ligand-binding domain, which is expected to render the VDR non-functional. Nevertheless, successful outpatient treatment was achieved with frequent high doses of oral calcium.
[52]
Kanakamani J, Tomar N, Kaushal E, et al. Presence of a deletion mutation (c.716delA) in the ligand binding domain of the vitamin D receptor in an Indian patient with vitamin D-dependent rickets type II[J]. Calcif Tissue Int, 2010, 86(1): 33-41. DOI: 10.1007/s00223-009-9310-2.
[53]
Linglart A, Biosse-Duplan M, Briot K, et al. Therapeutic management of hypophosphatemic rickets from infancy to adulthood[J]. Endocr Connect, 2014, 3(1): R13-R30. DOI: 10.1530/EC-13-0103.
In children, hypophosphatemic rickets (HR) is revealed by delayed walking, waddling gait, leg bowing, enlarged cartilages, bone pain, craniostenosis, spontaneous dental abscesses, and growth failure. If undiagnosed during childhood, patients with hypophosphatemia present with bone and/or joint pain, fractures, mineralization defects such as osteomalacia, entesopathy, severe dental anomalies, hearing loss, and fatigue. Healing rickets is the initial endpoint of treatment in children. Therapy aims at counteracting consequences of FGF23 excess, i.e. oral phosphorus supplementation with multiple daily intakes to compensate for renal phosphate wasting and active vitamin D analogs (alfacalcidol or calcitriol) to counter the 1,25-diOH-vitamin D deficiency. Corrective surgeries for residual leg bowing at the end of growth are occasionally performed. In absence of consensus regarding indications of the treatment in adults, it is generally accepted that medical treatment should be reinitiated (or maintained) in symptomatic patients to reduce pain, which may be due to bone microfractures and/or osteomalacia. In addition to the conventional treatment, optimal care of symptomatic patients requires pharmacological and non-pharmacological management of pain and joint stiffness, through appropriated rehabilitation. Much attention should be given to the dental and periodontal manifestations of HR. Besides vitamin D analogs and phosphate supplements that improve tooth mineralization, rigorous oral hygiene, active endodontic treatment of root abscesses and preventive protection of teeth surfaces are recommended. Current outcomes of this therapy are still not optimal, and therapies targeting the pathophysiology of the disease, i.e. FGF23 excess, are desirable. In this review, medical, dental, surgical, and contributions of various expertises to the treatment of HR are described, with an effort to highlight the importance of coordinated care.
[54]
Lecoq AL, Brandi ML, Linglart A, et al. Management of X-linked hypophosphatemia in adults[J]. Metabolism, 2020, 103S: 154049. DOI: 10.1016/j.metabol.2019.154049.
[55]
Imel EA, Glorieux FH, Whyte MP, et al. Burosumab versus conventional therapy in children with X-linked hypophosphataemia: a randomised, active-controlled, open-label, phase 3 trial[J]. Lancet, 2019, 393(10189): 2416-2427. DOI: 10.1016/S0140-6736(19)30654-3.
X-linked hypophosphataemia in children is characterised by elevated serum concentrations of fibroblast growth factor 23 (FGF23), hypophosphataemia, rickets, lower extremity bowing, and growth impairment. We compared the efficacy and safety of continuing conventional therapy, consisting of oral phosphate and active vitamin D, versus switching to burosumab, a fully human monoclonal antibody against FGF23, in paediatric X-linked hypophosphataemia.In this randomised, active-controlled, open-label, phase 3 trial at 16 clinical sites, we enrolled children with X-linked hypophosphataemia aged 1-12 years. Key eligibility criteria were a total Thacher rickets severity score of at least 2·0, fasting serum phosphorus lower than 0·97 mmol/L (3·0 mg/dL), confirmed PHEX (phosphate-regulating endopeptidase homolog, X-linked) mutation or variant of unknown significance in the patient or a family member with appropriate X-linked dominant inheritance, and receipt of conventional therapy for at least 6 consecutive months for children younger than 3 years or at least 12 consecutive months for children older than 3 years. Eligible patients were randomly assigned (1:1) to receive either subcutaneous burosumab starting at 0·8 mg/kg every 2 weeks (burosumab group) or conventional therapy prescribed by investigators (conventional therapy group). Both interventions lasted 64 weeks. The primary endpoint was change in rickets severity at week 40, assessed by the Radiographic Global Impression of Change global score. All patients who received at least one dose of treatment were included in the primary and safety analyses. The trial is registered with ClinicalTrials.gov, number NCT02915705.Recruitment took place between Aug 3, 2016, and May 8, 2017. Of 122 patients assessed, 61 were enrolled. Of these, 32 (18 girls, 14 boys) were randomly assigned to continue receiving conventional therapy and 29 (16 girls, 13 boys) to receive burosumab. For the primary endpoint at week 40, patients in the burosumab group had significantly greater improvement in Radiographic Global Impression of Change global score than did patients in the conventional therapy group (least squares mean +1·9 [SE 0·1] with burosumab vs +0·8 [0·1] with conventional therapy; difference 1·1, 95% CI 0·8-1·5; p<0·0001). Treatment-emergent adverse events considered possibly, probably, or definitely related to treatment by the investigator occurred more frequently with burosumab (17 [59%] of 29 patients in the burosumab group vs seven [22%] of 32 patients in the conventional therapy group). Three serious adverse events occurred in each group, all considered unrelated to treatment and resolved.Significantly greater clinical improvements were shown in rickets severity, growth, and biochemistries among children with X-linked hypophosphataemia treated with burosumab compared with those continuing conventional therapy.Ultragenyx Pharmaceutical and Kyowa Kirin International.Copyright © 2019 Elsevier Ltd. All rights reserved.
[56]
Insogna KL, Briot K, Imel EA, et al. A randomized, double-blind, placebo-controlled, phase 3 trial evaluating the efficacy of burosumab, an anti-FGF23 antibody, in adults with X-linked hypophosphatemia: week 24 primary analysis[J]. J Bone Miner Res, 2018, 33(8): 1383-1393. DOI: 10.1002/jbmr.3475.
In X-linked hypophosphatemia (XLH), inherited loss-of-function mutations in the PHEX gene cause excess circulating levels of fibroblast growth factor 23 (FGF23), leading to lifelong renal phosphate wasting and hypophosphatemia. Adults with XLH present with chronic musculoskeletal pain and stiffness, short stature, lower limb deformities, fractures, and pseudofractures due to osteomalacia, accelerated osteoarthritis, dental abscesses, and enthesopathy. Burosumab, a fully human monoclonal antibody, binds and inhibits FGF23 to correct hypophosphatemia. This report summarizes results from a double-blind, placebo-controlled, phase 3 trial of burosumab in symptomatic adults with XLH. Participants with hypophosphatemia and pain were assigned 1:1 to burosumab 1 mg/kg (n = 68) or placebo (n = 66) subcutaneously every 4 weeks (Q4W) and were comparable at baseline. Across midpoints of dosing intervals, 94.1% of burosumab-treated participants attained mean serum phosphate concentration above the lower limit of normal compared with 7.6% of those receiving placebo (p < 0.001). Burosumab significantly reduced the Western Ontario and the McMaster Universities Osteoarthritis Index (WOMAC) stiffness subscale compared with placebo (least squares [LS] mean ± standard error [SE] difference, -8.1 ± 3.24; p = 0.012). Reductions in WOMAC physical function subscale (-4.9 ± 2.48; p = 0.048) and Brief Pain Inventory worst pain (-0.5 ± 0.28; p = 0.092) did not achieve statistical significance after Hochberg multiplicity adjustment. At week 24, 43.1% (burosumab) and 7.7% (placebo) of baseline active fractures were fully healed; the odds of healed fracture in the burosumab group was 16.8-fold greater than that in the placebo group (p < 0.001). Biochemical markers of bone formation and resorption increased significantly from baseline with burosumab treatment compared with placebo. The safety profile of burosumab was similar to placebo. There were no treatment-related serious adverse events or meaningful changes from baseline in serum or urine calcium, intact parathyroid hormone, or nephrocalcinosis. These data support the conclusion that burosumab is a novel therapeutic addressing an important medical need in adults with XLH.© 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.© 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.
PDF(3892 KB)

2673

Accesses

0

Citation

Detail

Sections
Recommended

/